CA2078539C - Process for producing humanized chimera antibody - Google Patents

Process for producing humanized chimera antibody Download PDF

Info

Publication number
CA2078539C
CA2078539C CA002078539A CA2078539A CA2078539C CA 2078539 C CA2078539 C CA 2078539C CA 002078539 A CA002078539 A CA 002078539A CA 2078539 A CA2078539 A CA 2078539A CA 2078539 C CA2078539 C CA 2078539C
Authority
CA
Canada
Prior art keywords
antibody
variable region
dna
chain variable
heavy chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002078539A
Other languages
French (fr)
Other versions
CA2078539A1 (en
Inventor
Kenya Shitara
Nobuo Hanai
Mamoru Hasegawa
Hiromasa Miyaji
Yoshihisa Kuwana
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyowa Kirin Co Ltd
Original Assignee
Kyowa Hakko Kogyo Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kyowa Hakko Kogyo Co Ltd filed Critical Kyowa Hakko Kogyo Co Ltd
Publication of CA2078539A1 publication Critical patent/CA2078539A1/en
Application granted granted Critical
Publication of CA2078539C publication Critical patent/CA2078539C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated gangliosides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence

Abstract

A process for the production of humanized chimera antibody, wherein the chimera antibody is produced easily without changing any of the amino acids of its mouse antibody variable region, which comprises the steps of: (1) constructing a cassette vector by inserting a cDNA coding for a heavy chain constant region of human antibody into an expression vector for animal cell use and establishing a cloning site in the upstream region of the heavy chain constant region of said cassette vector for inserting a cDNA which encodes a heavy chain variable region of nonhuman animal antibody; (2) digesting a cDNA coding for the heavy chain variable region of nonhuman animal antibody with restriction enzymes; (3) inserting said cDNA coding for the heavy chain variable region of nonhuman animal antibody into the cassette vector, using a synthetic DNA
which comprises a base sequence corresponding to the 5'-end side of said heavy chain constant region of human antibody and a base sequence corresponding to the 3'-end side of said heavy chain variable region of nonhuman animal antibody and is possessed of the restriction enzyme recognition sites on both of its ends, thereby constructing a humanized chimera antibody heavy chain expression vector in which said cDNA coding for the heavy chain constant region of human antibody and said cDNA
coding for the heavy chain variable region of nonhuman animal antibody are linked together through said synthetic DNA; (4) constructing a cassette vector by inserting a cDNA coding for a light chain constant region of human antibody into an expression vector for animal cell use and establishing a cloning site in the upstream region of the light chain constant region of said cassette vector for inserting a cDNA which encodes a light chain variable region of nonhuman animal antibody; (5) digesting a cDNA coding for the light chain variable region of nonhuman animal antibody with restriction enzymes; (6) inserting said cDNA coding for a light chain variable region of nonhuman animal antibody into the cassette vector using a synthetic DNA which comprises a base sequence corresponding to the 5'-end side of said light chain constant region of human antibody and a base sequence corresponding to the 3'-end side of said light chain variable region of nonhuman animal antibody and is possessed of the restriction enzyme recognition sites on both of its ends, thereby constructing a humanized chimera antibody light chain expression vector in which said cDNA coding for the light chain constant region of human antibody and said cDNA coding for the light chain variable region of nonhuman animal antibody are linked together through said synthetic DNA; (7) introducing these expression vectors into host cells to obtain a transformant; and (8) culturing said transformant in an appropriate culture medium, thereby allowing the transformant to produce and accumulate a humanized chimera antibody, and collecting said humanized chimera antibody from the resulting culture broth.

Description

2~~~~3~
Process for Producing Humanized Chimera Antibody FIELD OF THE INVENTION
This invention relates to a process for the production of humanized chimera antibody.. In contrast to mouse monoclonal antibody, hurcianized chimera antibody. does not cause formation of anti-mouse immunoglobulin antibody in the body of a patient.
Thus, side effects are reduced or eliminated and half life in blood increases when the chimera antibody is used. Therapeutic effects which are superior to those obtained in the case of using mouse monoclonal antibody can be obtained in the treatment of human cancers and the like.
BACKGROUND OF THE INVENTION
It is known that, when mouse antibodies are administered to humans, they are recognized as foreign substances and cause formation of anti-mouse immunoglobulin antibodies in the human body, and the thus formed antibodies react with the administered mouse antibodies. As a result, side effects occur (J. Clin. Oncol., 2, 881 (1984); Blood, 65, 1349-1363 (1985); J. Natl. Cancer Ins~t., 80, 932 (1988); Proc.
Natl. Acad. Sci. U.S.A., 82, 1242 (1985)), the antibodies are cleared away quickly (J. Nucl. Med., 26, 1011 (1985); Blood, 65, 1349-1363 (1985); J. Natl. Cancer Inst., 80, 937 (1988)) and effects of the antibodies are reduced (J. Immunol., 135, 1530 (1985); Cancer Res., 4.6, 6489 (1986)). When mouse monoclonal antibody is converted into humanized chimera antibody, human anti-mouse immunoglobulin antibody form in _ 1 minimal amounts if at all, and the half life of the chimera antibody in human blood is six times as long as that of mouse monoclonal antibody (Proc. Natl. Acad. Sci. U.S.A., 86, 4220 ( 1989 ) ) . In addition, it is probable that the Fc region of mouse antibody does not fully activate human complement and human effector cells, in comparison with the Fc region of human antibody. For example, the antitumor activity of mouse monoclonal antibody to ganglioside GD2, which is effected via human effector cells, is improved when the monoclonal antibody is converted into chimera antibody that has the human antibody Fc region (J. Immunol., 144, 1382-1386 (1990)).
Ganglioside is one of the animal cell membrane-constituting glycolipids and is composed of a sugar chain as a hydrophilic side chain, sphingosine as a hydrophobic side chain and fatty acids. It is known that expression of ganglioside varies depending on the type of cells, organs and animal species. In addition, it has been revealed that quantity and quality of the expressed ganglioside change during the canceration process of cells (Cancer Res., 45, 2405 (1985)).
For example, it has been reporte-d 'that gangliosides GD2, GD3, GMZ and the like which hardly exist in normal cells were found in the cells of neuroblastoma, lung small cell carcinoma and melanoma belonging to neuroectodermal-origin tumor which is said to be highly malignant (J. Fxp. Med. , 155, 1133 ( 1982 ) ; J.
Biol. Chem., 257, 12752 (1982); Cancer Res., 47, 225 (1987);
ibid., 4?, 1098 (1987); ibid., 45, 2642 (1985); Proc. Natl.

~~"~~~~~
Acad. Sci. U.S.A., 80, 5392 (1983)).
Ganglioside GD3 has been found most frequently in melanoma cells among the neuroectodermal-origin tumors, and anti-ganglioside GD3 monoclonal antibodies (to be referred to as "anti-GD3 monoclonal antibody" hereinafter) belonging to the mouse IgM class and IgG class have been reported (Int. J.
Cancer, 29, 269 (1982); J. Biol. Chem., 257, 12752 (1982);
Cancer Res . , 47, 225 ( 1987 ) ; Acta Neuropathol . , 79, 317 ( 1989 ) ;
Proc. Natl. Acad. Sci. U.S.A., 77, 6114 (1980); J. Exp. Med., 155, 1133 (1982); Proc. Natl. Acad. Sci. U.S.A., 81, 5767 (1984)).
KM-641 (FERM BP-3116) disclosed in EP-A-0 493 686 is an anti-GD3 monoclonal antibody belonging to the mouse IgG3 class, which reacts not only with ganglioside GD3 but also with ganglioside 3',8'-LD1 and is possessed of a broad range of antitumor spectrum. In addition, KM-641 has stronger binding activities to antigens than anti-GDS monoclonal antibody R24 which has been disclosed in J. Exp. Med., 155, 1133 (1982) and it shows strong antitumor activities.
The mouse monoclonal antibody R24 to the gangliaside GD3 was once used for the treatment of melanoma, but the administered mouse monoclonal antibody R24 did not fully exert its effect due to the formation of anti-mouse immunoglobulin antibody in the patient's body (Eur. J. Cancer Clin. Oncol., 24, suppl 2, s 65 (1988)).
Consequently, the use of chimera antibody for anti-GD3 monoclonal antibody would be advantageous in that anti-mouse immunoglobulin antibody does not form in the body, side effects are reduced or eliminated, its half life in blood is prolonged and its antitumor effector effect increases, and thus therapeutic effects of the chimera antibody which axe superior to 'those of mouse monoclonal antibody can be obtained in the treatment of human cancers and the like.
Several processes for the production of humanized chimera antibodies are known. Humanized chimera antibody, in which constant regions of the heavy chain (to be referred to as "H chain" hereinafter) and the light chain (to be referred to as "L chain" hereinafter) of mouse monoclonal antibody are converted into human constant regions, is produced in animal cells making use of recombinant DNA techniques. Examples of such processes include a process in which humanized chimera antibody is produced using chromosomal DNA as a gene which encodes mouse H chain variable region (to be referred to as "VH" hereinafter) and L chain variable region (to be referred to as "VL" hereinafter) (Morrison et al., Proc. Natl. Acad.
Sci. U.S.A., 81, 6851 (1984); Neuberger et al., Nature, 314, 268 (1985); Nishimura et al., Cancer Res., 47, 999 (1987);
Dorai et al., J. Immunol., 139, 4232 (1987); Kameyama et al., FEBS letter, 244, 301 (1989)) and another process in which humanized chimera antibody is produced using cDNA (Gillies et al., J. Immunol. Methods, 125, 191 (1989); Liu et al., published International Application in Japan No. 2-501886).
_ 4 _ ~~"~~~~~' Cloning and base sequence determination of hybridoma cell chromosomal DNA which encodes mouse Vx and VL require much time and labor in comparison with those of cDNA that encodes mouse VH and VL. Consequently, the process in which cDNA is used for the production of humanized chimera antibody is more desirable than the chromosomal DNA process.
Gillies et al. have succeeded in expressing humanized chimera antibody in animal cells, making use of an expression vector for animal cells having inserted therein a humanized chimera H chain gene obtained by linking mouse VH-encoding cDNA
with human CH-encoding chromosomal DNA, arid a humanized chimera L chain gene obtained by linking mouse VL-encoding cDNA with human CL-encoding chromosomal DNA (J. Immunol. Methods, 125, 191 (1989)). However, when an attempt was made to prepare chimera antibodies from several types of antibodies, a problem was found that there were certain chimera antibodies whose L
chains could not be expressed without converting leader sequences. In additian, humanized chimera antibody can be produced more simply when cDNA which encodes human C,~ and CL is used instead of the human CH- and CL-encoding chromosomal DNA.
In published International Application in Japan No. 2-501886, Liu et al. discloses a process for the expression of humanized chimera antibody in animal cells, which comprises using an expression vector for animal cells having inserted therein a chimera H chain cDNA obtained by linking mouse V~-encoding cDNA with human Cg-encoding cDNA and a chimera L chain _ ~~''~
cDNA obtained by linking mouse VL-encoding cDNA with human CL-encoding cDNA. According to this process, however, it is necessary to alter the JH portion of the VH-encoding cDNA and the JL portion of the VL-encoding cDNA by means of mutation, because the cDNA which encodes mouse VH or VL is linked with the human CH- or CL-encoding cDNA at the J region in the mouse variable region. In addition, with regard to the chimera L
chain prepared using mouse Jk5, leucine which is one of the amino acids of the framework 4 is changed to isoleucine when made into humanized chimera antibody. Although amino acid sequence of complementarity-determining region (to be referred to as "CDR" hereinafter) is especially important for antigen-antibody binding, the amino acid sequence of the framework is also an important factor. Fox example, Riechmann et al. have prepared CDR graft antibody by grafting a rat antibody CDR into a human antibody framework and reported that binding activity of the antibody was reduced by the framework conversion and the antibody activity increased when amino acid sequence of the framework was partially changed (Nature, 332, 323 (1988)).
Consequently, there is a possibility that the binding activity of humanized chimera antibody is undesirably reduced when the antibody is produced by the mouse Jk5-aided process disclosed by Liu et al.
In view of the above, when any mouse antibody is converted iota humanized chimera antibody, it has been desired to simply and easily produce humanized chimera antibody in which amino acids of the mouse antibody variable region remain completely unchanged.
OBJECTS OF THE INVENTION
An object of the present invention is to provide a process for the production of humanized chimera antibody by which the chimera antibody is produced easily without changing any of the amino acids of its mouse antibody variable region.
Another object of the present invention is to provide a humanized. chimera antibody to ganglioside GD3 and a process for the production of such antibody.
' SUMMARY OF THE INVENTION
The present invention relates to a process for producing humanized chimera antibody which comprises the steps of (1) constructing a cassette vector by inserting a cDNA coding for human antibody CH into an expression vector for animal cell use and establishing a cloning site in the upstream region C~
of said cassette vector for inserting a cDNA which encodes nonhuman animal VH;
(2) digesting a cDNA coding for nonhuman animal antibody VA
with restriction enzymes;
(3) inserting said cDNA coding for nonhuman animal antibody VH into the cassette vector, using a synthetic DNA
which comprises a base sequence corresponding to the 5'-end side of said human antibody CH, for example, the 31$' to 43rd nucleotide in SEQ ID No: 7, and a base sequence corresponding to the 3'-end side of said nonhuman animal antibody VH and is possessed of restriction enzyme recognition sites on both of its ends, thereby constructing a humanized chimera antibody H chain expression vector in which said cDNA coding for human antibody CH and said cDNA coding for nonhuman animal antibody VH are linked together through said synthetic DNA;
(4)~constructing a cassette vector by inserting a cDNA coding for human antibody CL into an expression vector for animal cell use and establishing a cloning site in the upstream region of the CL of said cassette vector for inserting a cDNA which encodes nonhuman animal antibody VL;
( 5 ) digesting a cDNA coding for nonhuman animal antibody VL
with restriction enzymes;
(6) inserting said cDNA coding for nonhuman animal antibody VL
into the cassette vector, using a synthetic DNA which comprises a base sequence corresponding to 'the 5'-end side of said human antibody CL and a base sequence corresponding to the 3'-end side of said nonhuman animal antibody VL and is possessed of restriction enzyme recognition sites on both of its ends, thereby constructing a humanized chimera antibody h chain expression vector in which said cDNA coding for human antibody CL and said cDNA coding for nonhuman animal antibody VL are linked together through said synthetic DNA;
(7) introducing these expression vectors into host cells to obtain a transformant; and (8) culturing said transformant in an appropriate culture medium, thereby allowing the transformant to. produce and _ 8 _ accumulate a humanized chimera antibody, and collecting said humanized chimera antibody from the resulting culture broth.
The cassette vector to be used in the present invention is a vector which is obtained by inserting a~cDNA that encodes a constant region of human antibody into an expression vector fox animal cell use, in which a cloning site is located in the upstream region of the constant region for inserting a cDNA
that encodes a variable region of nonhuman animal antibody. An expression vector for humanized chimera antibody can be constructed easily by inserting a variable region of nonhuman animal antibody into the cloning site of the cassette vector, using a synthetic DNA which comprises a base sequence corresponding to the 5'-end side of a constant region of human antibody and a base sequence corresponding to the 3'-end side of a variable region of nonhuman animal antibody and is possessed of restriction enzyme recognition sites on its both ends.
The present invention also relates to a humanized chimera antibody obtainable by the above-described process, a pharmaceutical composition comprising the h~,unanized chimera antibody and a pharmaceutically acceptable carrier, and use of said humanized chimera antibody for the manufacture of'the medicament for treating cancer.
BRIEF DESCRTPTION OF THE DRAWINGS
Fig. 1 shows a restriction enzyme cleavage map of a 9.3 kb XbaT fragment of KM50 cell chromosomal DNA.
_ g 2~~~~ a~
Fig. 2 shows a construction scheme for plasmi.d pKMBll.
Fig. 3 shows a construction scheme for plasmid pKMD6.
Fig. 4 shows a construction scheme for plasmid pEPKMAl.
Fig . 5 shows a construction scheme for plasmid pEPKMBl .
Fig. 6 shows a construction scheme for plasmid pAGE501.
Fig. 7 shows a construction scheme for plasmid pAGE109.
Fig. 8 shows a construction scheme for plasmid pAGE502.
Fig > 9 shows a construction scheme for plasmid pAGE503 .
Fig. 10 shows a construction scheme for plasmid pSEdl.
Fig. 11 shows a construction scheme for plasmid pSElD2.
Fig. 12 shows a~ construction scheme for plasmid pIGISEId2.
Fig. 13 shows a construct~.on scheme ror plasmia pIGISEld3.
Fig. 14 shows a construction scheme for plasmid pIGISEld4.
Fig. 15 shows a construction scheme for plasmid pPMOL2.
Fig. 15 shows a construction scheme far plasmid pFM0L3.
Fig. 17 shows a construction scheme for plasmid pchCKA7.
Fig. 18 shows a construction scheme for plasmid pchCKBl.
Fig. 19 shows a construction scheme for plasmid pckCKCl.
Fig. 20 shows a construction scheme for plasmid pChi Ig~-IB2 .

2~
Fig. 21 shows a construction scheme for plasmid pChiIgLAl.
Fig. 22 shows plasmids plCM641HA3 and pKM641LA2.
Fig. 23 shows plasmid pChi641HA1.
Fig. 24 shows a construction scheme for plasmid pICM641HE1 .
Fig. 25 shows a construction scheme for plasmid pKM641HF1.
Fig. 26 shows a construction scheme for plasmitl pChi641HA1.
Fig. 27 shows a construction scheme for plasmid pChi641HAMl.
Fig. 28 shows plasmid pChi641LG11.
Fig. 29 shows a construction scheme for plasmid pChi641LG11~.
Fig. 30 shows a construction scheme for plasmid pChi641LGM11.
Fig. 31 shows a pattern of SDS-PAGE (4 to 155 gradient gel) of purified anti-GD3 chimera antibody KM-871~(about 5 ug/lane) carried out under reductive condition (A) or non-reductive condition (B), where the lanes starting from the left respectively indicate electrophoretic patterns of molecular weight markers, human IgG standard, mouse anti-GD3 antibody KM-641 and anti-GD3 chimera antibody KM-871.
Fig. 32 is a graph showing reactivity of anti-GD3 chimera antibody KM-871 with ganglioside GD3-positive 6361 and 2~~~ ~ ~~~
SK-MEL-28 cells measured by fluorescent antibody technique with the cell number on the ordinate and the fluorescence intensity on the abscissa. A dotted line shows reactivity in the absence of the antibody, while a solid line shows reactivity in the presence of KM-871.
Fig. 33 is a graph showing complement-dependent cytotoxicity (CDC) of anti-GD3 chimera antibody KM-871 and anti-GD3 mouse antibody KM-641 against ganglioside GD3-positive 6361 and SK-MEL-28 cells with cytotoxicity on the ordinate and an antibody concentration on the abscissa. A blackened bar shows CDC activity of KM-871, while a striped bar shows that of KM-641.
Fig. 34 is a graph showing antibody-dependent cell-mediated cytotoxicity (ADCC) of KM-871 and KM-641 against ganglioside GD3-positive cell 6361 with a ratio of effector cells to target cells on the ordinate and ADCC activity on the abscissa. A blackened bar shows ADCC activity of KM-871, a dotted bar shows ADCC activity of KM-641 and a striped bar shows control (in the absence of the antibody). PMN means polymorphonuclear leukocyte.
Fig. 35 is a graph showing therapeutic effect of KM-871 on transplanted tumors with the tumor size on the ordinate'and days after transplantation of tumors on the abscissa, in which closed circle shows effect of anti--Sialyl Lea monoclonal antibody AMC-462, open square shows that of KM-641 and open triangle shows that of KM-871.

~~~1~~~~
DETAILED DESCRIPTION OF THE INVENTION
I. Construction of cassette vector The cassette vector to be used in the present invention is constructed by inserting a cDNA which encodes a human antibody constant region into an expression vector for animal cell use. Essential components in the expression vector for animal cell use include promoter, enhancer, polyA signal, splicing signal, drug resistance gene as a selection marker (e.g., ampicillin resistance gene, etc.) and the like. Any expression vector for animal cell use may be used for this purpose, as long as it can contain and express the cDNA
molecule which encodes a human antibody constant region. For example, pAGE107 (Cytotechnology, 3, 133 (1990)) is useful as such an expression vector. Examples of the promoter and enhancer for use in the expression vector for animal cell use include: Sv40 early promoter and enhancer (J. Biochem., 101, 1307 (1987)); LTR promoter and enhancer of Moloney mouse leukemia virus (Biochem. Biophys. Res. Comun., 149, 960 (1987)); and immunoglobulin H chain promoter (Cell, 41, 479 (1985)) and enhancer (Cell, 33, 717 (1983)). The immunoglobulin H chain promoter and enhancer can be prepared using appropriate antibody-producing hybridoma cells, such as rat hybridoma KM50 cells which produce anti-human serum albumin antibody as disclosed in JP-A-60-258128 (the term "JP-A" as used herein means an "unexamined published Japanese patent application"). The following describes processes for the ~° D~'~,rt preparation of the immunoglobulin H chain promoter and enhancer making use of KM50 cells.
Each chromosomal DNA is obtained from cultured KM50 cells, anal P3X63Ag8U.1 (to be referred to as "P3U1"
hereinafter) cells (ATCC CRL1597) which are to be fused with KM50 and rat kidney cells in accordance with the procedure disclosed in Molecular Cloning (2nd. ed., Cold Spring Harbor Laboratory Press, 1989, p9.14). Next, a DNA fragment containing immunoglobulin promoter and enhancer and a gene of the variable region of activated immunoglobulin H chain, in which DNA rearrangement has been induced, is isolated from the chromosomal DNA extracted from KM50 cells, in accordance with the procedure disclosed in FEBS letter 244, 301 (1989). The immunoglobulin promoter and enhancer axe cut out from the thus isolated DNA fragment and inserted into the aforementioned expression vector for animal cell use. Plasmid pTglSEld4 is an illustrative example of the animal cell expression vector which contains the immunoglobulin H chain promoter and enhancer.
Next, a cloning site is established in the upstream region of a human constant region of a cassette vector, for inserting a cDNA which encodes a variable region of nonhuman animal antibody. Into the thus established cloning site is inserted a cDNA which encodes a variable region of nonhuman animal antibody, using a synthetic DNA which comprises a base sequence corresponding to the 5'-end side of a constant region of human antibody and a base sequence corresponding to the 3'-end side of a variable region of nonhuman animal antibody and is possessed of restriction enzyme recognition sites on both of its ends . In this way, a humanized chimera antibody expression vec~.or is constructed in which the cDNA coding for human antibody constant region and the cDNA coding fox the variable region of nonhuman animal antibody are linked-together through the synthetic DNA. The synthetic DNA to be used may be prepared using a DNA synthesizer, based on the base sequence which corresponds to the 5'-end side of a constant region of human antibody and the base sequence that corresponds to the 3'-end side of a variable region of nonhuman animal antibody.
Illustrative examples of cloning site-containing cassette vectors include a cassette vector pChiIgHB2 which is used for the construction of an expression vector for 'the expression of humanized chimera antibody H chain and a cassette vector pChiIgLA2 which is used for the canstruction of an expression vector for the expression of humanized chimera antibody L
chain.
A cassette vector for use in the construction of an expression vector for the expression of humanized chimera antibody H chain is constructed, for example, by cutting out a human C~-encoding cDNA-containing fragment, from an ApaI site in the vicinity of the 5'-end of the cDNA to its 3'-end, and inserting the fragment into an appropriate expression vector for animal cell use such as plasmid pzglSHld4 or the like.
Then, a cloning site is established in the thus constructed cassette vector for inserting a cDNA which encodes a VH of nonhuman animal antibody. Into the thus established cloning site is then inserted a cDNA fragment encoding a nonhuman animal antibody VH, which is obtained by digesting a VH-encoding cDNA with an appropriate restriction enzyme, using a synthetic DNA'molecule which comprises a base sequence corresponding to the 5'-end side (5'-end to ApaI site) of a human antibody CH
and a base sequence corresponding to the 3'-end side of a nonhuman animal antibody V~ and is possessed of restriction enzyme recognition sites on both of its ends. In this way, an express~.on vector for use in the expression of humanized chimera antibody H chain is easily obtained without altering amino acid sequence of the expressed V~.
A cassette vector for constructing of an expression vector for the expression of humanized chimera antibody L chain may be constructed for example by introducing an EcoRV site into the vicinity of 5' -end side of a human CL-encoding cDNA by means of mutation, cutting out a fragment from the resulting human cDNA from the EcoRV site to the 3'-end and inserting the fragment into an appropriate expression vector such as plasmid pIglSEld4 or the like. Then, a cloning site is established in the thus constructed cassette vector for inserting a cDNA which encodes a nonhuman animal antibody VL. Into the thus established cloning site is then inserted a cDNA fragment encoding a nonhuman animal antibody V~,, which is obtained by digesting a VL-encoding cDNA with an appropriate restriction enzyme, using a synthetic DNA which comprises a base sequence corresponding to the.5'-end side (5'-end to EcoRV site) of a human antibody CL and a base sequence corresponding to the 3'-end side of a nonhuman animal antibody VL and is possessed of restriction enzyme recognition sites on both of its ends. In 'this way, an expression vector for use in the expression of humanized chimera antibody L chain is easily obtained without altering amino acid sequence of the expressed VL.
Examples of the cDNAs which encode the human CH and human CL described above are disclosed, for instance, in Cell 22, 197 (1982). Such eDNAs can be prepared from human antibody-producing myeloma cells, humanized monoclonal antibody-producing.hybridoma cells, humanized chimera antibody-producing cells (SP2-PC chimera; FEBS Letters, 244, 301 ( 1989 ) ) and the like, in accordance with known procedures disclosed for instance in Proc. Natl. Acad. Sci. U.S.A. 82, 7025 (1985) and ibid., 79 7025 (1985) . That is, cDNA is synthesized using mRNA extracted from the above-described cells, in accordance with the procedure disclosed in Molecular Cloning 2nd. ed.;
1989, p8.1. A library is prepared from the thus synthesized cDNA using a phage vector or a plasmid vector, in accordance , with the procedure disclosed in Molecular Cloning 2nd. ed.;
1989, p8.1, 1.53. Next, a recombinant phage or a recombinant plasmid which contains human C~-encoding cDNA or human CL-encoding cDNA is obtained from the thus prepared library using a human antibody constant region or a human antibody variable region as a probe, in accordance with the procedure disclosed in Molecular Cloning 2nd. ed.; 1989, p8.1, 1.53. Base sequences of the human CH-encoding cDNA and the human CL-encoding cDNA are determined in accordance with the procedure disclosed in Molecular Cloning, 2nd. ed.; 1989, p13.1.
Tntxoduc~tion o.f an appropriate restriction enzyme recognition site into the human CL-encoding cDNA, for example insertion of an EcoRV recognition site into a region in the vicinity of the 5'-end o~ the cDNA, maybe effected in accordance with the procedure disclosed in Molecular Cloning, 2nd. ed.; 1989, p15.1.
2. Production of humanized chimera antibody Firstly, cDNAs which encode VH and VL of nonhuman animal antibody, such as mouse anti-GD3 monoclonal antibody, are prepared in the .following manner.
That is, cDNA is synthesized using mRNA extracted from appropriate hybridoma cells which produce mouse anti--GD3 monoclonal antibody, such as mouse anti-GDS monoclonal antibody KM-641 (FERM BP-3116). A library is prepared from the thus synthesized cDNA using a phage vector or a plasmid vector.
Next, a recombinant phage or a recombinant plasmid which contains VA-encoding cDNA or VL-encoding cDNA is obtained -from the thus prepared library using a constant region or a variable region of nonhuman antibody, such as mouse antibody, as a probe. Base sequences of the V~-encoding cDNA and the VL-encoding cDNA are determined in accordance with the ~~~'~~~3-aforementioned procedure.
A fragment of the VH-encoding cDNA, ranging from the 5'-end to an appropriate restriction enzyme site near the 3'-end (to be referred to as "site A" hereinafter), is cut out and inserted into the cloning site of the aforementioned cassette vector, using a synthetic DNA which comprises a base sequence corresponding to the 5'-end side of a human antibody CH and a base sequence corresponding to the 3.'-end side (from 3'-end to site A) of a nonhuman animal antibody VH and is possessed of restriction enzyme recognition sites on both of its ends. In this way, an expression vector for use in the expression of humanized chimera antibody H chain is constructed by linking the human antibody Ca-encoding cDNA with the nonhuman antibody VH-encoding cDNA trrough the synthetic DNA. In the same way, a fragment of the VL-encoding cDNA, ranging from the 5'-end to an appropriate restrictian enzyme site near the 3'-end (to be referred to as "site B" hereinafter), is cut out and inserted into the cloning site of the aforementioned cassette vector, using a synthetic DNA molecule which comprises a base sequence corresponding to the 5'-end side of a human antibody C~, and a base sequence corresponding to the 3'-end side (from 3'-end to site B) of a nonhuman animal antibody V~, and is possessed of restriction enzyme recognition sites on both of its ends. In this way, an expression vector for use in the expression of humanized chimera antibody h chain is constructed by linking the human antibody CL-encoding cDNA with the nonhuman antibody 2~~~~'~Y
VL-encoding cDNA through the synthetic DNA.
A transformant which is capable of producing humanized chimera antibody is obtained by transforming appropriate host cells with the thus prepared expression vectors for use in the expression of the H chain and L chain of humanized chimera antibody.
Any type of cells may be used as host cells for use in the introduction of the humanized chimera antibody expression vectors, as long as these cells are capable of expressing the humanized chimera antibody. Illustrative examples of such host cells include mouse SP2/0-Agl4 cells (ATCC CRL1581; to be referred to as "SP2/0 cells" hereinafter), mouse P3X63-Ag8.653 (ATCC CRL1580) and CHO cells which are deficient in dihydrofolate reductase gene (to be referred to as "dhfr"
hereinafter) (Urlaub et al., Proc. Natl. Acad. Sci~. U.S.A., 77, 4216 (1980)).
Introduction of the expression vectors for use in the expression of the H chain and L chain of humanized chimera antibody into host cells may be effected for example by the electroporation technique disclosed in JP-A-2-257891. A
transformant capable of producing the humanized chimera antibody may be selected using RPMI1640 medium supplemented with 6418 and fetal calf serum, in accordance with the procedure disclosed in JP-A-2-257891. A transformant, KM-871, which produces humanized chimera antibody that reacts with ganglioside GD3 is an illustrative example of the transformant "1~~~~.~
capable of producing humanized chimera antibody. KM-871 has been deposited on August 13, 1991, with Fermentation Research Institute, Agency of Industrial Science and Technology of 1-3, Higashi 1-chome, Tsukuba-shi, Ibaraki, Japan under the Budapest Treaty, and has been assigned the accession number FERM BP-3512.
For the cultivation of the thus-obtained transformants, any medium can be used as long as the desired antibody can be produced~and accumulated in the medium. An example of such medium is RPMI1640 medium supplemented with 6418 and fetal calf serum. The transfornnants may be inoculated into 200 ul to 100 ml of the above-mentioned medium to give a cell concentration of 1 x 105 to 1 x 10' cells/ml and cultivated at 37°C in a 5~
COZ incubator for 1 to 7 days . The desired chimera antibody is produced and accumulated in the culture medium.
Activity of the humanized chimera antibody in the culture broth is measured by enzyme-linked immunosorbent assay (ELISA method; E. Harlaw et ,al., Manual of Antibody Experiments, Cold Spring Harbor Laboratory Press, 1988).
Productivity of the humanized chimera antibody in the transformant can be improved making use of a dhfr amplification system in accordance with the procedure disclosed in JP-A-2-257891.
The humanized chimera antibody thus produced can be purified from supernatant fluid of the aforementioned cultured mixture making use of a protein A column { E . Harlow et al . , Manual of Antibody Experiments, Cold Spring Harbor Laboratory Press, 1988). Illustrative examples of humanized chimera antibodies obtained in this way include those which react with ganglioside GD3, such as humanized chimera antibody KM-871 and the like.
Reactivity of humanized chimera antibody is measured by ELISA method. The molecular weight of the H chain, the L chain or the entire molecule of purified humanized chimera antibody is measured by means of polyacrylamide gel electrophoresis (SDS-PAGE), Western blotting method (E. Harlow et al., Manual of Antibody Experiments, Cold Spring Harbor Laboratory Press, 1988) or the like.
Binding activity, or avidity, of the humanized chimera antibody to ganglioside GD3 to a cultured cancer cell line is measured by means of the fluorescent antibody technique, the ELISA method or the like. Complement-dependent cytotoxicity (CDC activity} and antibody-dependent cell-mediated cytotoxicity (ADCC activity) of humanized chimera antibody to a cultured cancer cell line are measured in accordance with the procedures disclosed in Menekigaku Jikken Nyumon, (Manual of Immunological Experiments) Matsuhashi et al., Gakkai Shuppan Center, Japan, 1981).
The humanized chimera antibodies according to the present invention can be used alone as an anticancer agent.
For example, the humanized chimera antibodies according to the present invention can be used in a pharmaceutical composition with a pharmaceutically acceptable carrier. They may be formulated into an anticancer composition together with at least one pharmaceutically acceptable carrier. For instance, the humanized chimera antibodies are dissolved in physiological saline, an aqueous solution of glucose, lactose or mannitol and the like. The powder of the humanized chimera antibodies for injection can be prepared by lyophilizing the humanized chimera antibodies in accordance with the conventional method and mixing the lyophilized products with sodium chloride. The anticancer composition may further contain additives conventionally used well known in the art of medical preparation, for example, pharmaceutically acceptable salts.
The humanized chimera antibodies according to the present invention can be administered in the form of the above-described anticancer composition to mammals including human in a dose of 0.2 to 20 mg/kg/day. The dose may vary depending on the age, condition, etc. of patients. The administration of the anticancer composition can be effected by intraveous injection once a day (single adm5.nistration or consecutive administration) or intermittently one to three times a week or once every two to three weeks.
The antincancer composition is expected to be useful for treating cancer such as melanoma, neuroblastoma and glioma.
The following examples and reference examples are provided to further illustrate the present invention. It is to be understood, however, that the examples are for purpose of illustration only and are not to be construed to limit the invention.

Example 1 Construction of cassette vector 1. Isolation of promoter and enhancer genes of KM50 cell-derived immunoglobulin H chain (1) Preparation of chromosomal DNA from KM50 cells, P3U1 cells and'rat kidney Chromosomal DNA was prepared in the following manner in accordance with the procedure disclosed in, Molecular Cloning, Maniatis et al., 1989, p9.16.
1.2 x 10$ KM50 cells, 2 x 108 P3U1 cells (ATCC CRL1597) and 1.6 g of rat kidney (a kidney sample frozen at -80°C was smashed thoroughly using a mallet) were each suspended in 2 ml of a buffer solution (pH 7.5) containing 10 mM Tris-HC1, 150 mM
sodium chloride and 10 mM sodium ethylenediaminetetraacetate (to be referred to as "EDTA" hereinafter). To this suspension were added 0.8 mg of Proteinase K (Sigma Chemical Co.) and 10 mg of sodium lauryl sulfate (to be referred to as "SDS"
hereinafter). After,incubation at 37°C for 10 hours, the resulting mixture was extracted with the same volume of phenol (once), chloroform (twice) and ether (once) in this order, and the extract was dialyzed for 10 hours against a buffer solution (pH 7.5) containing 10 mM Tris-HC1 and 1 mM EDTA. A DNA
solution was recovered from the dialysis tube and Ribonuclease A (Sigma Chemical Co.) was added thereto to give a final concentration of 20 ug/ml. After incubating at 37°C for 6 hours to decompose RNA completely, the resulting solution was mixed with 15 mg of SDS and 1 mg of Proteinase K and incubated at 37°C for 10 hours. The thus treated solution was extracted with the same volume of phenol, chloroform and ether (twice for each) in this order, and the extract was dialyzed for 10 hours against 'a buffer solution (pH 7.5) containing of 10 mM Tris-HC1 and~1 mM EDTA. The DNA solution was recovered from the dialysis tube and used as a chromosomal DNA sample. A DNA
concentration of each sample was determined by measuring the absorbance at 260 iun and, as a result, it was found 'that 1.6 mg, 1.5 mg and 1.9 mg of chromosomal DNA was obtained from 1.2 x 108 KM50 cells, 2 x 108 P3U1 cells and 1.6 g of rat kidney, respectively.
(2) Identification of activated immunoglobulin ~t chain gene in KM50 cells by Southern blotting A 3 ug portion of each of the chromosomal DNA samples obtained in the above step (1) from KM50 cells, P3U1 cells and rat kidney was dissolved in 25 ul of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mrI magnesium chloride and 100 mM sodium chloride. Each of the thus prepared solution was mixed with 15 units of XbaT (Takara Shuzo Co., Ltd.; all restriction enzymes used in the following experiments were purchased from the same company) and incubated at 37°C for 2 hours to cleave the chromosomal DNA at the Xbal site. The reaction mixture was subjected to agarose gel electrophoresis, resulting DNA fragments were transferred to a nitrocellulose filter in accordance with the method of Southern et al. (J.

Mol. Biol., 98, 503, (1975)) and then subjected to hybridization in the known method (Kameyama et al., FEBS
Letters, 244, 301-306 (1989)) using a mouse JH probe which is disclosed in the FEBS Letters article. A band equivalent to about 9 . 3 kb was observed only in the DNA sample of KM50 cells .
In consequence, it was considered that the XbaI fragment of immunoglobulin DNA found in this band contained the activated immunoglobulin H chain gene derived from KM50 cells.
(3) Preparation of KM50 cell chromosomal DNA library A 60 ug portion of the KM50 cell chromosomal DNA
obtained in the above step (2) was dissolved in 250 ~1 of a buffer solution containing lO mM Tris-HCl (pH 7.5), 6 mM
magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 150 units of Xbal and incubated at 37°C for 2 hours to cleave the chromosomal DNA at 'the XbaI site. The reaction mixture was subjected to agarose gel electrophoresis and a 9.3 kb-equivalent fraction was recovered as about 2 ~g of 9.3 kb DNA sample of KM50 cells, making use of the DEAE paper method (Maniatis et al., Molecular Cloning, 1989, p6.24). Separately, a 3 ug portion of lambda-ZAP ( Stratagene Cloning Systems ) to be used as a vector was dissolved in 200 ~1 of a buffer solution containing 10 mM Tris-HCl (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 50 units of XbaI and incubated at 37°C for 2 hours to cleave the DNA at the XbaT site. The resulting reaction mixture was extracted with phenol-chloroform and then treated with ethanol to precipitate and recover about 3 ~g of DNA. The thus recovered DNA sample was dissolved in a 100 ~1 of 100 mM Tris-HCl buffer (pH 7.5), and the resulting solution was mixed with 1 unit of alkaline phosphatase (Takara Shuzo Co., Ltd.) to effect dephosphorylation of restriction enzyme cleavage ends of the vector DNA. The resulting reaction mixture was extracted with phenol-chloroform and then treated with ethanol to precipitate and recover 2 ug of DNA. The thus recovered DNA sample was dissolved in 10 ~l of a buffer solution containing 10 mM Tris-HC1 (pH 7.5) and 1 mM EDTA to serve as a vector DNA sample.
Next, 0.2 ug of the thus prepared vector DNA sample and 0.2 ~g of the KM50 cell-derived 9.3 kb DNA sample were dissolved in 5 ~1 of a buffer solution containing 66 mM Tris-HC1 (pH 7.5), 6.6 mM magnesium chloride, 10 mM dithiothreitol (to be referred to as "DTT" hereinafter) and 0.1 mM adenosine triphosphate (to be referred to as "ATP" hereinafter)(to be referred to as "T4 ligase buffer" hereinafter). The resulting solution was mixed with 175 units of T4 DNA ligase (Takara Shuzo Co., Ltd.) and incubated at 4°C for 3 days. A 2 ~1 portion of the resulting reaction mixture was subjected to lambda phage packaging in the known method (Maniatis et al., Molecular Cloning, 1989, p2.95) using GigaPackTM Gold purchased from Stratagene Cloning Systems.
E. coli BB4 cells were infected with this phage to obtain 200,000 phage clones. 100,000 out of these phage clones were fixed on nitrocellulose filters in the known method (Maniatis et al., Molecular Cloning, 1989, p2.112).
(4) Selection of recombinant DNA containing a gene of the activated (anti-human serum albumin) immunoglobulin H chain variable region in KM50 cells Two clones showing strong reaction with the 32P-labeled mouse JH probe at 65°C were isolated from the 100,000 phage clones prepared in the above step (3) in accordance with the procedure of Kameyama et al. (FEBS Letters, 44, 301-306, 1989 ) .
When the~phage DNA was recovered in the conventional manner (Maniatis et al., Molecular Cloning, 1989, p2.118-2.169), it was found that the 9.3 kb XbaI fragment of the KM50 cell-derived chromosomal DNA was incorporated into the phage DNA.
(5) Base sequence of the gene of the activated (anti-human serum albumin) immunoglobulin H chain variable region in KM50 cells Restriction enzyme cleavage maps of the two clones obtained in the above step (4) was prepared by digesting them with various restriction enzymes and it was found that completely the same DNA fragment ( 9 . 3 kb) has been inserted into these clones (Fig. 1). Next, base sequence of a part of the 9.3 kb DNA fragment:, which was considered to contain the promoter and variable regions of the rat immunoglobulin H
chain, was determined in accordance with the Banger method (Banger et al., Proc. Natl. Acad. Sci. U.S.A., 74, 5463 {1977);
M13 Cloning and Sequencing Handbook, Amersham). In SEQ ID N0:
1, a region containing octamer sequences such as ATGCAAAT and ~~rl ~~~
TATA box sequences such as TTGAAAA and the like can be regarded as 'the immunoglobulin promoter region.
2. Construction of heterologous protein expression vector using promoter and enhancer of the activated (anti-human serum albtunin) immunoglobulin H chain variable region in KM50 cells (1)~Construction of pKMBl1 A 1 ~g portion of the 9.3 kb fragment of the 'immunoglobulin H chain variable region gene obtained in 1-(5) was dissolved in 30 ul of a buffer solution containing 10 mM
Tris-HC1 (pH 7.5), s mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of BalII and 10 units of HindIII and incubated at 37°C for 2 hours to cleave the DNA fragment at the BalII and HindIII
sites. The resulting reaction mixture was subjected to agarose gel electrophoresis and 0.01 ~g of a DNA fragment containing 0.8 kb immunoglobulin promoter was recovered. Separately, a 1 ug portion of a plasmid pBR322-BalIT (Kuwana et al., FEBS
Letters, 219, 360 (187)) was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of Bc~lII and 10 units of HindIII and incubated at 37 °C for 2 hours to cleave the plasmid at the BaIII and HindIII sites. The resulting reaction mixture was subjected to agarose gel electrophoresis, a DNA fragment of about 4.2 kb was recovered. A 0.1 ug portion of the thus obtained pBR322-BalTI derived DNA fragment of about 4.2 kb and 0.01 ug of the immunoglobulin promoter-containing DNA fragment were dissolved in 20 ul of a T4 ligase buffer, and the resulting solution was mixed with 175 units of T4 DNA ligase (Takara Shuzo Co., Ltd.) and incubated at 4°C for 24 hours.
Using the .resulting reaction mixture, transformation of E. coli T-TB101 (J. Mol. Biol., 41, 459 (1969)) was carried out in accordance with the method of Scott et al. (M.Shigesada, Saibo Kogaku, 2, 616 (1983)) to isolate a colony having ampicillin resistance (to be referred to as "ApR" hereinafter). Plasmid DNA was recovered from the colony to obtain pKMBll as shown in Fig. 2.
(2) Construction of pKMD6 In order to establish an appropriate restriction enzyme recognition site in downstream region of the immunoglobulin ' promoter, the plasmid pKMBll constructed in the above step (1) was digested with nuclease BAL31 from the NcoI site. A 10 ~g portion of the plasmid pKMBIl was dissolved in 100 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM
magnesium chloride and 50 mM potassium chloride. The thus prepared solution was mixed with 30. units of NcoT and incubated at 37°C fox 2 hours to cleave the plasmid at the NcoI site.
The resulting reaction mixture was extracted with phenol and chloroform and treated with ethanol. The thus precipitated DNA
fragments were dissolved in 100 ~l of BAL31 buffer which contained 20 mM Tris-HC1 (pH 8.0), 600 mM sodium chloride, 12 mM calcium chloride, 12 mM magnesium chloride and 1 mM EDTA, and the resulting solution was mixed with 0.25 unit of BAL31 (Bethesda Research Laboratories, Inc. (BRL)) and incubated at 37 °C for 5 seconds . The reaction was stopped by extracting the reaction mixture with phenol. After extraction with chloroform and precipitation with ethanol, 1 ~g of DNA was recovered. A
0.1 ~g portion of the thus obtained DNA sample and 0.01 ~g of a synthetic DNA linker Sall were dissolved in 20 ~1 of the T4 lipase buffer, and the resulting solution was mixed with 175 units of T4 DNA lipase and incubated at 4°C for 24 hours.
Using the resulting reaction mixture, transformation of E. coli HB101 was carried out in accordance with the method of Scott et al. to isolate an ApR colony. Plasmid DNA was recovered from the colony to obtain pKMD6 as shown in Fig. 3. The base sequence of the BAL31-digested portion of this plasmid was determined in accordance with the Sanger method and it was found that bases up to the third base (the 303 position base in the SEQ ID N0: 1 ) upstream from the initiation colon ATG of the immunoglobulin gene.
(3) Construction of pEPKMA1, pEPKMBl and pAGE501 Since original promoter and enhancer of the immunoglobulin gene are separated from each other, it is necessary to construct a vector in which the promoter~and enhancer are connected together so that it can be used as a vector for the expression of a heterolagous protein. The following manipulation was carried out to construct such vectors.

D~;K~~~
~~'~~~.~~:~~3 A 1 wg portion of the 9.3 kb fragment of the .
immunoglobulin H chain variable region gene obtained in 1-(5) was dissolved in 30 ~1 of a buffer solution containing 10 mM
Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The 'thus prepared solution was mixed with 10 units of EcoRV and 10 units of XbaI and incubated at 37°C for 2 hours to cleave the DNA fragment at the EcoRV and XbaI sites. The resulting reaction mixture was subjected to agarose gel electrophoresis and 0.1_ ~g of a DNA fragment of about 1 kb containing the immunoglobulin enhancer region was recovered.
Separately, a 1 ug portion of the plasmid pKMD6 obtained in the above step (2) was dissolved in 100 ~l of a buffer solution containing 10 mM Tris-HC1 ( pH 7 . 5 ) , 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of BalII and incubated at 37°C for 2 hours to cleave the plasmid at the salIl site. The resulting reaction mixture was extracted with phenol and chloroform and precipitated with ethanol. The thus precipitated DNA fragments were dissolved in 40 ul of DNA polymerase I buffer containing 50 mM Tris-HC1 (pH 7.5), 10 mM magnesium chloride, 0.1 mM dATP
(deoxyadenosine triphosphate), 0.1 mM dCTP (deoxycytidine triphosphate), 0.1 mM dGTP (deoxyguanosine triphosphate) and 0.1 mM dTTP (deoxythymidine triphosphate). The resulting solution was mixed with 5 units of E . coli DNA polymerase I
Klenow fragment and incubated at 16°C for 90 minutes to convert the cohesive 5'-end formed by the actllI digestion into blunt end. The reaction was stopped by extracting the reaction mixture with: phenol. After extraction with chloroform and precipitation with ethanol, the resulting DNA fragments were dissolved in 30 ul of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and incubated at 37°C for 2 hours to cleave the DNA
fragment at the HindTII site. The resulting reaction mixture was subjected to agarose gel electrophoresis, 0.1 ug of a DNA
fragment of about 0.8 kb containing the immunoglobulin promoter region was recovered. Next, a 0.2 ug portion of plasmid pUCl8 (Messing, Methods in Enzymology, 101, 20 (1983)) was dissolved in 30 ~xl of a buffer solution containing 10 mM Tris-HC1 (pH
7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and units of XbaI and incubated at 37°C for 2 hours to cleave the plasmid at the HindIII and Xbal sites. The resulting reaction mixture was subjected to agarose gel electrophoresis, 0.1 ug of a DNA fragment of about 2.7 kb was recovered. A 0.1 ug portion of the thus obtained pKMD6-derived 0.8 kb DNA
fragment, 0.02 ug of the DNA fragment containing the immunoglobulin enhancer region and 0.1 ug of the pUCl8 fragment were dissolved in 20 ~1 of the T4 lipase buffer, and the resulting solution was mixed with 175 units of T4 DNA lipase and incubated at 4°C for 24 hours. Using the resulting reaction mixture, transformation of E. coli HB101 was carried ~~~1~~~~
out to isolate an ApR colony. Plasmid DNA was recovered from the colony to obtain pEPKMAl as shown in Fig. 4.
Next, a 1 ug portion of the plasmid pEPKMAl was dissolved in 100 ~.1 of a buffer solution containing 10 mM Tris-HG1 (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chlc5ri.de. The thus prepared solution was mixed with 10 units of Xbal and incubated at 37°C for 2 hours to cleave the plasmid at the Xbal site. The resulting reaction mixture was extracted with phenol and chloroform and precipitated with ethanol. The thus precipitated DNA fragments were dissolved in 40 ~1 of the aforemetioned DNA polymerise I buffer solution, and the resulting solution was mixed with 6 units of E. coli DNA
polymerise I Klenow fragment and incubated at 16°C for 90 minutes to convert the cohesive 5'-end formed by the XbaI
digestion into blunt end. The reaction was stopped by extracting the reaction mixture with phenol. After extraction with chloroform and precipitation with ethanol, DNA fragments was recovered. The thus obtained DNA sample and 0.01 ~g of a synthetic DNA XhoI linker (Takara Shuzo Co., Ltd.) were dissolved in 20 ul of the T4 ligase buffer, and the resulting solution was mixed with 175 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the resulting reaction mixture, transformation of E. coli HB101 was carried out to isolate an ApR colony. Plasmid DNA was recovered from the colony to obtain pEPKMBl as shown in Fig. 5.
Next, SV40 early gene promoter and enhancer regions (to 2~'~~~ ~~
be referred to as "PSE" hereinafter) of an expression vector pAGE107 for use in the expression of heterologous genes in animal cells (Miyaji et al., Cytotechnology, 3, 133 - 140 ( 1990 ) ) were converted into KM50-derived immunoglobulin H chain promoter and enhancer (to be referred to as "PxH" hereinafter) of pEPICMB1 in the following manner.
A 1 ~,g portion of the plasmid pAGE107 was dissolved in 30 ~tl of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 150 mM sodium chloride. The thus prepared solution was mixed with 10 units of SalI and 10 units of XhoI and incubated at 37 °C for 2 hours to cleave the plasmid at the SalI and XhoI sites . The resulting reaction mixture was subjected to agarose gel electrophoresis and 0.5 ug of a DNA
fragment of about 5.95 kb containing 6418 resistance gene was recovered. Next, a 1 ~g portion of the plasmid pEPKMBl was dissolved in 30 ul of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and :150 mM sodium chloride. The thus prepared solution was mixed with 10 units of SalI and 10 units of Xhol and incubated at 37°C for 2 hours to cleave the plasmid at the SaII and XhoI sites. The resulting reaction mixture was subjected to agarose gel electrophoresis and 0.1 ~g of a DNA fragment of about 1.7 kb containing immunoglobulin promoter and enhancer regions was recovered. A 0.1 fag portion of the thus obtained pAGE107-derived 5.95 kb DNA fragment and 0.02 ~g of the DNA fragment containing immunoglobulin promoter and enhancer regions were ~~~1~~~'~
dissolved in 20 ul of the T4 ligase buffer, and the resulting solution was mixed with 175~ units of T4 DNA ligase and incubated at 4°C for 24 hours. Using 'the resulting reaction mixture, transformation of E, coli HB101 was carried out to isolate an ApR colony. Plasmid DNA was recovered from the colony to obtain pAGE501 as shown in Fig. 6.
(4) Construction of pAGE109 ' One of the two EcoRI cleavage sites in plasmid pAGE106 was deleted in the following manner to construct pAGE109.
A 2 ug portion of the expression vector pAGE106 for use in the expression.of heterologous genes in animal cells (JP-A
3-22979 or EP-A-0 405 285) was dissolved in 100 ul of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of SacI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1.5 ~g of a pAGE106 DNA fragment (4.3 kb) was recovered which contained the SV40 early gene promoter and 6418 resistance gene cleaved with EcoRI and Sacl. The thus recovered DNA fragment was dissolved in 40 ul of the DNA polymerise I buffer solution, and the resulting solution was mixed with 5 units of E. coli DNA polymerise I large fragment and incubated at 16°C for 2 hours to convert the cohesive 3'-end formed by the SacI
digestian and the cohesive 5'-end formed by the EcoRI digestion into blunt ends. The resulting reaction mixture was extracted ., with phenol and chloroform and then treated with ethanol. The thus precip5.tated sample was dissolved in 20 ~1 of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 4 hours. Using the thus obtained recombinant plasmid DNA, transformation of E.
coli HB101 was carried out to obtain plasmid pAGE109 as shown in Fig. 7.
(5) Construction of pAGE502 Plasmid pAGE502 was constructed in the following manner in order to convert the SV40 promoter and enhancer of pAGE107 into immunoglobulin H chain promoter and enhar_cer.
A 2 ~g portion of the plasmid pAGE107 disclosed in JP-A-3-22979 or EP-A-0 405 285 was dissolved in 100 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of ~IindIII and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation and the thus recovered sample was dissolved in 40 ul of the DNA polymerase I buffer solution. The resulting solution was mixed with 5 units of E. coli DNA polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the HindIII digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of XhoI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1.5 ~g of a pAGE107 DNA fragment of about 5.95 kb was obtained which contained 6418 resistance gene and ApR gene cleaved with XhoI and HindIII.
Next, a 2 ~g portion of the plasmid pAGE501 obtained in the above. step ( 3 ) was dissolved in 100 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 175 mM sodium chloride. The thus prepared solution was mixed with 10 units of Sall and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ~1 of the DNA polymerase I buffer solution.
The resulting solution was mixed with 5 units of E. coli DNA
polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the SaII digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ul of a buffer solution containing ZO mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of XhoI and incubated at 37°C
for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis arid about 0.2 ug of a pAGE501 DNA
~g _ fragment of about 1.8 kb was obtained which contained KM50 immunoglobulin H chair. promoter and enhancer genes cleaved with XhoI and SalI.
Next, 0.1 ~g of the thus obtained pAGE107 HindIII-XhoI
fragment (about 5.95 kb) and 0.1 ~g of the pAGE501 SalI-XhoI
fragment (about 1.8 kb) were dissolved in 20 ~1 of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours.
Using the.thus obtained recombinant plasmid DNA, transformation of E, coli HB101 was carried out to obtain plasmid pAGE502 as shown in Fig. 8.
(6) Construction of pAGE503 One of the two EcoRI cleavage sites in plasmid pAGE502 was deleted in the following manner to construct pAGE503.
A 2 ~g portion of the plasmid pAGE109 obtained in the above step ( 4 ) was dissolved in 30 ~l of a buffer solution containing 10 mM Tris-HCl (pH 7 . 5 ) , 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and 10 units of ClaI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.2 ug of a pAGE109 DNA fragment of about 1 kb was recovered which contained the poly (A) signal gene of beta-globin and SV40 early genes cleaved with ClaI and HindITI.
Next, a 2 ~g portion of the plasmid pAGE502 obtained in the above step (5) was dissolved in 30 ~.1 of a buffer solution s containing 10 mM Tris-HC1 (pH 7 , 5 ) , 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and 10 units of Clal and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and then to the aforementioned DEAF paper method to recover about 1 ~g of a pAGE502 DNA
fragment of about 6.l~kb which contained KM50 immunoglobulin H
chain promoter and enhancer genes, ApR gene and 6418 resistance gene cleaved with HindIII and ClaI. Next, 0.1 ug of the thus obtained pAGE109 HindIII-ClaI fragment (about 1 kb) and 0.1 ~g of the pAGE502 HindIII-Clal fragment {about 6.1 kb) were dissolved in 20 ~xl.of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C far 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain plasmid pAGE503 as shown in Fig. 9.
(7) Construction of pSEld1 A dhfr gene was introduced,into plasmid pAGE107 in the following manner to construct plasmid pSEldl.
A 2 ug portion of the plasmid pAGE107 disclosed in JP-A
3-22979 or EP-A-0 405 285 was dissolved in 100 ~1 of a buffer solution containing 100 mM Tris-HC1 (pH 7.5), 5 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRT and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the ~~'~8~~~
thus recovered sample was dissolved in 40 wl of the DNA
polymerase I buffer solution. The resulting solution was mixed with 5 units.of E. coli DNA polymerase T Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the EcoRI digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ~l of a buffer solution which was composed of mM Tris-HCl ( pH 7 . 5 ) , 6 mM magnesium chloride and 50 mM
sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1.5 ~g of a pAGE107 DNA fragment of about 5. 6 kb was recovered which contained 6418 resistance gene and ApR gene cleaved with EcoRI and HindIII.
Next, a 2 ~g portion of a plasmid pSV2-dhfr (Subramani et al . , Mol . Cell . Biology, 1, 854 ( 1981 ) ) was dissolved in 100 ul of a buffer solution containing 10 mM Tris-HCl (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with ZO units of BalII and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ~1 of the DNA polymerase I buffer solution. The resulting ' salution was mixed with 5 units of E. coli DNA polymerase I
Klenow fragment and incubated at 16°C for 2 hours to convert ~~p~C3vJ
the cohesive 5'-end formed by the BctlII digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 100'mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and incubated at 37°C for 4 hours.
The resulting reaction mixture was subjected to agarose gel electrophoresis, about 0.2 ~g of a pSV2-dhfr DNA fragment of about 0.76 kb was recovered which contained dhfr gene cleaved with BalII and HindIII.
Next, 0.1 ~g of the thus obtained pAGE107 HindIII-EcoRI
fragment (about 5.6 kb) and 0.1 ug of the pSV2-dhfr B~lII-HindIII fragment (about 0.76 kb) were dissolved in 20 dal of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours.
Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain plasmid pSEldl as shown in Fig. 10.
(8) Construction of pSEld2 The HindIII cleavage site was removed from the plasmid pSEld1 in the following manner to construct plasmid pSEld2.
A 2 ug portion of the plasmid pSEld1 obtained in the above step (7) was dissolved in 100 ul of :a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 7.0 units of HindIII and incubated at 37 °C for 4 hours .
After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 pl of the DNA polymerase :L buffer solution. The resulting solution was mixed with 5 units of E. 0017. DNA polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the I-IindIII digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 20 ~l of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E~ coli HB101 was carried out to obtain plasmid pSEld2 as shown in Fig. 11.
(9) Construction of pIglSEld2 The dhfr gene was introduced into plasmid pAGE503 in the following manner to construct plasmid pIglSEld2.
A 2 ug portion of the plasmid pAGE503 obtained in the above step (6) was dissolved in 100 ~l of a buffer solution containing 100 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of Clal and incubated at 37°C for 4 hours.
After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ~1 of the DNA polymerase ~~~~'D3 I buffer solution. The resulting solution was mixed with 5 units of E. coli DNA polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the Clal digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HCl (pH 7.5), 6 mM magnesium chloride and 50 mM sodium chloride.. The thus prepared solution was mixed with 10 units of MluI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1 ~g of a pAGE503 DNA fragment of about 5.4 kb was recovered which contained the KM50 immunoglobulin H chain promoter and enhancer genes cleaved with Clal and MluI.
Next, a 2 ~.g portion of the plasmid pSEld~ obtained in the above step ( 8 ) was dissolved in 100 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7 . 5 ) , 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with. 10 units of XhoI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation and the thus recovered sample was dissolved in 40 ul of the DNA polymerase I buffer solution. The resulting solution was mixed with 5 units of E.
coli DNA polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the Xhol digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 30 ~.1 of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM
magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of Mlu2 and incubated at 37°C fox 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1 ug of a pSEld2 DNA fragment of about 3.8 kb was recovered which contained.dhfr gene cleaved with XhoI and Mlu2.
Next, 1 ~g of the thus obtained pAGE503 ClaI-MluI
fragment (about 5.4 kb) and 1 ug of the pSEld2 XhoI-MluI
fragment (about 3.8 kb) were dissolved in 20 ~1 of the T4 lipase buffer, and the resulting solution was mixed with 350 units of T4 DNA lipase and incubated at 4°C for 24 hours.
Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain plasmid pIglSEld2 as shown in ~'ig . 12 .
( 10 ) Construct:ion of pIglSEld3 The ApaI cleavage site was removed from the plasmid pIglSEld2 in the following manner to construct plasmid pIglSEld3.
A 2 ug portion of the plasmid pIglSEld2 obtained in the above step ( 9 ) vas dissolved in 100 ul of a buffer solution containing 10 mM Tris-HCl (pH 7.S) and 6 mM magnesium chloride.
The thus prepared solution was mixed with 10 units of ApaI and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ul of the DNA polymerase I buffer solution. The resulting solution was mixed with 5 units of E. coli DNA polymerase I
Klenow fragment and incubated at 16°C for 2 hours to convert the'cohesive 3'-end formed by the ApaI digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 20 ~1 of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E.
coli HB101 was carried out to obtain plasmid pIglSEld3 as shown in Fig. 13.
(11) Construction of pTglSEld4 In order to establish a cloning site between HindIII
cleavage site and EcoRI cleavage site of the plasmid pIglSEld3, plasmid pIglSEld4 was constructed by inserting the synthetic DNA shown in SEQ ID N0:2 into the plasmid pIglSEld3 in the following manner.
A 2 ~g portion of the plasmid pIglSEld3 obtained in the above step (10) was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 ( pH 7 . 5 ) , 5 mM magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of HindIII and 10 units of EcoRI and incubated at 37 °C for 4 hours . The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1 ug of a pIglSEld3 ANA fragment of about 9.2 kb was recovered which contained the KM50 immunoglobulin H chain' promoter and enhancer genes, ApR
gene, 6418 resistance gene and dhfr gene cleaved with HindIII
and EcoRI.
Next, 0.1 ~g of the thus obtained pIglSEld3 HindIII-EcoRI fragment (about 9.2 kb) and 10 ng of the synthetic DNA
(SEQ ID N0: 2) were dissolved in 20 ul of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA
ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E, coli HB101 was carried out to obtain plasmid pTglSEld4 as shown in Fig. 14.
3. Preparation of Moloney mouse leukemia virus long terminal repeat (to be referred to as "MoLTR" hereinafter) Since MoLTR is known to have promoter and enhancer activities (Kuwana et al., Biochem. Biophys. Res. Comun., 149, 960 (1987)), a plasmid pPMOL3 containing MoLTR was prepared in the following manner in order to use MoLTR as cassette vector promoter and enhancer.
A 3 ug portion of the plasmid pPMOLI disclosed in JP-A
1-63394 was dissolved in 30 ~l of a buffer solution containing mM Tris-HCl (pH 7.5), 7 mM magnesium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of CIaI and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ul of the DNA polymerase I buffer solution.
The resulting solution was mixed with 5 units of E. coli DNA
polymerase I Klenow fragment and incubated at 16°C for 2 hours to convert the cohesive 5'-end formed by the ClaI digestion into blunt end. The reaction was stopped by phenol extraction, followed by chloroform extraction and ethanol precipitation to recover 2 ~g of DNA fragments. The thus precipitated DNA
sample and 0.01 ug of a synthetic DNA XhoI linker (Takara Shuzo Co. , Ltd. ) were dissolved in 20 ~1 of the T4 lipase buffer, and the resulting solution was mixed with 175 units of T4 DNA
lipase and incubated at 4°C for 24 hours. Using the resulting reaction mixture, transformation of ~. cola HB101 was carried out to obtain plasmid pPMOL2 as shown in Fig. 15. Next, a 3 ~.g portion of the thus obtained plasmid ppMOL2 was dissolved in 30 ~l of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 7 mM magnesium chloride, 10 mM sodium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of Smal and incubated at 37 °C for 4 hours . The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation and 2 ~g of DNA fragments were recovered. The thus recovered DNA sample and 0.01 ug of a synthetic DNA EcoRI linker (Takara Shuzo Co., Ltd.) were dissolved in 20 ~1 of the, T4 lipase buffer, and the resulting solution was mixed with 175 units of T4 DNA lipase and incubated at 4°C for 24 hours. Using the resulting reaction _ 48 mixture, transformation of E. coli HB101 was carried out to obtain plasmid pPMOL3 as shown in Fig. 16.
4. Cloning of H chain constant region (Cgl) cDNA and L chain constant region (Ck) cDNA of human immunoglobulin IgGl (1) Preparation of mRNA from chimera antibody-producing SP2-PC
Chimera-1 cells Using a mRNA extraction kit, Fast TrackT~' (No. K1593-02, available from Invitrogen), 6.2 ~g of mRNA was obtained from 1 x 10g cells of chimera antibody-producing SP2-PC Chimera-1 which has anti-phosphorylcholine activity and is disclosed in FEBS Letters (244, 301 - 306 (1989)).
( 2 ) Preparation of SP2-PC Chimera-1 cDNA library and cloning of human immunoglobulin H chain constant region (Cgl) cDNA and L
chain constant region (Ck) cDNA
A 2 ~g portion of the mRNA obtained in the above step (1) was subjected to EcoRI adaptor addition using cDNA
Synthesis Kit (No. 27-9260-Ol, available from Pharmacia) followed by kination. The resulting cDNA solution was subjected to phenol-chloroform extraction and ethanol precipitation to recover 4 ~g of cDNA. The thus recovered cDNA
was dissolved in 20 ~l of sterile water, and the resulting solution was subjected to agarose gel electrophoresis to recover about 0.3 ~g of a DNA fragment of about 1.8 kb and about 0.3 ~g of a DNA fragment of about 1.0 kb.
Next, a 5 ~g portion of the vector pUCl8 was dissolved in 100 ~1 of a buffer solution containing 100 mM Tris-HC1 (pH

~~"~°.~
7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 50 units of EcoRI and incubated at 37°C for 4 hours to cleave the pUCl8 DNA at its EcoRI cleavage site. The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation to recover about 3 ug of a pUCl8 DNA fragment cleaved with EcoRI.
Next, 0.1 ~g of the thus obtained pUCl8 EcoRI fragment ( about 2 . 7 kb ) and the 1. 8 kb and 1. 0 kb cDNA fragments ( 0 .1 ~g for each) prepared from the SP2-PC Chimera-1 cells were dissolved in 20 ~z7. of the T4 ligase buffer, and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours.
Using the thus obtained recombinant plasmid DNA, transformation of E. coli LE392 was carried out. About 3,000 colonies 'thus obtained were fixed on nitrocellulose filters.
Two 32P-labeled probes were prepared from human immunoglobulin constant region chromosomal genes (Cgl as an IgGl H chain constant region and Ck as an IgG1 L chain constant region) which have been isolated by Kameyama et al. (FEBS Letters, 244, 301 (1989)). From colonies which showed strong reactions at 65°C with these probes, one showing,strong reaction with Cgl (pPCVHhCGII) and the other showing strong reaction with Ck (pPCVLhCKl) were obtained.
(3) Introduction of EcoRV site into human Ig k chain constant region An EcoRV site was introduced into 5'-end side of the human Ig k chain constant region by means of site-specific mutagenesis using a kit purchased from Promega (Catalogue No.
Q6210). A 2 ~g portion of the plasmid pPCVLhCKl was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HCl (pH
7.5), 6 mM magnesium chloride and 5U mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of Kpn,I and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.2 ug of a pPCVLhCKl DNA fragment of about 0.8 kb was recovered which contained the human immunoglobulin L chain constant region cleaved with EcoRI and KpnI.
Next, a 2 ~.g portion of .pSELECTl (a kit available from Promega, Catalogue No. Q6210) was dissolved in 30 ul of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 6 mM
magnesium chloride and 50 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of KpnI arid incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1 ug of a pSELECT1 DNA fragment of about 5.7 kb cleaved with EcoRI and KpnI was recovered.
Next, 0.1 ~g of the pPCVLhCKl EcoRI-Kpnl fragment (about 0.8 kb) and 0.1 wg of the pSELECTl EcoR2-Kpnl fragment (about 5.7 kb) obtained above were dissolved in 20 ul of the T4 lipase buffer, and the resulting solution was mixed with 350 units of T4 DNA lipase and incubated at 4°C for 24 hours.

~~~(D3J ~~
Using the thus obtained recombinant plasmid DNA, transformation of E. coli JM109 was carried out to obtain plasmid pchCKA7 as shown in Fig. 17.
Next, using the plasmid pchCKA7 thus obtained and the synthetic DNA of SEQ ID N0:3 as mutagenesis primer, the ACC
sequence of the human immunoglobulin L chain constant region (12 to 1~ position bases from the N-terminal) was converted into GAT in order to construct a plasmid pchCKBl (Fig. 18) in which an ~EcoRV site was introduced into the converted site.
Next, the EcoRV site of the plasmid pchCKB1 was converted into HindIII cleavage site in the following manner.
A 2 ~g portion of the plasmid pchCKBl obtained above was dissolved in 10 dal of a buffer solution containing 100 mM Tris-HC1 (pH 7.5), 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRI and incubated at 37°C for 4 hours. After subjecting the resulting reaction mixture to phenol-chloroform extraction and ethanol precipitation, the thus recovered sample was dissolved in 40 ~1 of the DNA polymerase I buffer solution.
The resulting solution was mixed with 5 units of E. coli DNA
polymerase I Klenow fragment and incubated at 37°C for 30 minutes to convert the cohesive 5'-end formed by the EcoRI
digestion into blunt end. The resulting reaction mixture was extracted with phenol and chloroform and then treated with ethanol. The thus precipitated sample was dissolved in 20 ~1 of the T4 ligase buffer containing 0.1 ~g of HindIII linker (Takara Shuzo Co., Ltd.), and the resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain plasmid pchCKC1 as shown in Fig. 19.
5. Construction of cassette vector (1) Construction of a cassette vector for use in the construction of humanized chimera antibody H chain expression vector A 2 ~g portion of the plasmid pTgISEId4 obtained in the aforementioned step 2-(11) was dissolved in 30 ul of a buffer solution containing 10 mM Tris-HCl (pH 7 . 5 ) , 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRV and 10 units of ApaI
and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1.5 ~g of a pIgISEId4 DNA fragment of about 9.2 kb cleaved with EcoRV and Apal was recovered.
Next, a 2 ug portion of the plasmid pPCVHhCGII obtained in the aforementioned step 4-(2) was dissolved in 30 ~l of a buffer solution containing 10 mM Tris-HCl (pH 7.5) and 6 mM
magnesium chloride. The thus prepared solution was mixed with units of Apal and IO units of SmaI and incubated at 37°C for 1 hour. The resulting reaction mixture was subjeced to agarose gel electrophoresis and about 0.2 ug of a pPCVHhCGIl DNA
fragment of about 1 kb was recovered which contained the human immunoglobulin H chain constant -region gene cleaved with ApaI
and SmaI.
Next, 0.1 ~g of the pIgISEId4 EcoRV-ApaI fragment (about 9.2 kb) and 0.1 ~g of the pPCVHhCGII Apal-SmaI fragment (about 1 kb) prepared above were dissolved in 20 ~7. of the T4 ligase buffer. The resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E.
coli HB101 was carried out to obtain a plasmid pChiIgHB2 (Fig.
20) as a cassette vector for use in the construction of a humanized chimera antibody H chain expression vector.
(2) Construction of a cassette vector for use in the construction of humanized chimera antibody L chain expression vector A 2 ~g portion of the plasmid pIgISEId4 obtained in the aforementioned step 2-(11) was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HCl (pH 7 . 5 ) , 6 mM magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRV and 10 units of HindIII and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 1.5 ~g of a pIgISEId4 DNA fragment of about g.2 kb cleaved with EcoRV and HindIII was recovered.
Next, a 2 ug portion of the plasmid pchCKCl obtained in the aforementioned step 4-(3) was dissolved in 30 ul of a buffer solution containing ZO mM Tris-HC1 (pH 7.5), 6 m~I

magnesium chloride and 100 mM sodium chloride. The thus prepared solution was mixed with 10 units of EcoRV and 10 units of HindIII and incubated at 37°C for 1 hour. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.2 ~g of a pPCVLhCKl DNA fragment of about 0.6 kb was recovered which contained the human immunoglobulin L chain constant region gene cleaved with EcoRV and HindIII.
Next, 0.1 ug of the pIgISEId4 EcoRV-HindIII fragment ( about 9 ..2 kb ) and 0 . 1 ug of the pchCKC 1 EcoRV-Hind I I I f ragment (about 0.6 kb) prepared above were dissolved in 20 ~1 of the T4 ligase buffer. The resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E.
coli HB101 was carried out to obtain a plasmid pChiIgLAl (Fig.
21) as a cassette vector for use in the construction of a humanized chimera antibody L chain expression vector.
Example 2 Anti-GD3 chimera antibody 1. Preparation of mRNA from a hybridoma cell line capable of producing mouse anti-GD3 monoclonal antibody KM-641 Using a mRNA extraction kit, Fast Track''°'' (No. K1593-02, available from Invitrogen), 34 ug of mRNA was prepared from 1 x 108 cells of a hybridoma cell line which is capable of producing mouse anti-GD3 monoclonal antibody KM-641 prepared by the method described later in Reference Example shown below.
2. Preparation of cDNA libraries of KM-641 H chain and L chain genes Using ZAP-cDNA Synthesis Kit (No. sc200400), a cDNA
synthesis kit available from Stratagene Cloning Systems, cDNA
having EcoRI adaptor on its 5'-end and XhoI adaptor on its 3'-end was prepared from 3 ~g of the mRNA obtained in the above procedure 1. About 6 ~g of the cDNA was dissolved in 10 ~l of sterile water and subjected to agarose gel electrophoresis to recover 0.1 ~g of an H chain-corresponding cDNA fragment of about 1.8 kb and 0.1 ~g of an L chain-corresponding cDNA
fragment of about 1.0 kb. Next, 0.1 ug of the 1.8 kb cDNA
fragment, 0.1 ug of the 1.0 kb cDNA fragment and 1 ~g of Uni-ZAP XR (available from Stratagene Cloning Systems; a preparation obtained by digesting Lambda ZAPII vector with EcoRI and XhoI, followed by treatment with calf intestine alkaline phosphatase) to be used as a vector were dissolved in 11.5 ul of the T4 ligase buffer, and the resulting solution was mixed with 175 units of T4 DNA ligase and incubated at 12°C for hours and then at room temperature for 2 hours. A 4 ul portion of the resulting reaction mixture was subjected to lambda phage packaging using Giga PackTM Gold (Stratagene Cloning Systems) in accordance with the conventional method (Maniatis et al., Molecular Cloning, 1989, p2.95). An E. coli strain PLK-F was infected with the thus packaged product in accordance with the conventional method (Maniatis et al., Molecular Cloning, 1989, p2. 95-107 ) to obtain an H chain cDNA library and an L chain cDNA library, each containing about 10,000 phage clones. Next, these phage particles were fixed on nitrocellulose filters in accordance with the conventional method (Maniatis et al., Molecular Cloning, 1989, p2.112).
3. Cloning of monoclonal antibody KM-641 H chain and L chain cDNA
Two 'zP-labeled probes were prepared from an EcoRI
fragment of about 6.8 kb containing a mouse immunoglobulin constant region chromosomal gene Cg1 (Roeder et al., Proc.
Natl. Acad. Sci. U.S.A., 78, 474 (1981)) and a mouse Ck gene-containing Hindl I I-BamF-II fragment of about 3 kb ( Sakano et al . , Nature, 280, 288 (19x9)). A phage clone which showed strong reaction at 65°C with one of these two probes were obtained from each of 'the H chain cDNA library and the L chain cDNA
library prepared in the above procedure 2 in accordance with the conventional method (Maniatis et al., Molecular Cloning, 1989, p2.108). Next, using ZAP-cDNA Synthesis Kit (No.
sc200400), a cDNA synthesis kit of Stratagene Cloning Systems each of the thus obtained phage Clones was introduced into plasmid pBluescript to isolate a recombinant plasmid pKM641HA3 containing the KM-641 H chain cDNA and a recombinant plasmid pKM641LA2 containing the ICM-641 L chain cDNA. When each of the plasmids pKM641HA3 and pKM641LA2 was digested with EcoRI and XhoI, it was found that cDNA of about 1.6 kb had been inserted into the former plasmid, and cDNA of about 0.9 kb into the latter (Fig. 22).
4. Immunoglobulin variable region base sequences of KM-641 H

~~~Je,~c/
chain cDNA (pKM641HA3) and KM-641 L chain cDNA (pKM641LA2) Immunoglobulin variable region base sequences of the plasmids pKM641HA3 and pKM641LA2 obtained in the above procedure 3 were determined by the dideoxy method (Maniatis et al., Molecular Cloning, 1989, p13.42) using Sequenase Version 2.0'DNA Sequencing Kit (United States Biochemical Corporation).
The results are shown in SEQ ID N0:4 and SEQ ID N0:5. The plasmid pKM641LA2 was a complete cDNA containing a leader sequence and having a methionine-corresponding sequence which was assumed to be the initiation colon ATG located close to the 5'-end. The plasmid pKM641HA3, on the other hand, did not have such a methionine-corresponding initiation colon-like sequence on its 5'-end side, and its leader sequence was partially deficient.
5. Construction of KM-641 chimera H chain expression vector H chain variable region gene obtained by cleaving the plasmid pKM641HA3 variable region at the 5'-end Alul site and 3'-end Styl site was ligated with the cassette vector for use in the construction of the humanized chimera antibody H chain obtained in Example 1 using the synthetic DNA sequences shown in SEQ ID N0:6 and SEQ ID N0:7, thereby constructing a humanized chimera antibody H chain expression vector pchi641HA1 (Fig. 23).
Firstly, the DNA shown in SEQ ID N0: 7 ( see F'ig . 23 ) was synthesized using a DNA synthesizer. This synthetic DNA
comprises a base sequence derived from plasmid pKM641HA3 _ 58 -ranging from the 3'-end of its immunoglobulin H chain variable region to a StyI cleavage site in the vicinity of the 3'-end and a base sequence derived from plasmid pAGE28 ranging from the 5'-end of its immunoglobulin H chain constant region to an ApaI cleavage site in 'the vicinity of the 5'-end. Thus, the synthetic DNA has a StyI cleavage site and an ApaI cleavage site on both of its end. Next, the thus synthesized DNA was introduced into the plasmid pKM641HA3 in the following manner.
A 3 ~g portion of the plasmid pKM641HA3 was dissolved in 30 ~l of a buffer solution containing 50 mM Tris-HC1 (pH
7.5), 10 mM magnesium chloride, 50 mM sodium chloride and 1 mM
DTT. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of SCI and incubated at 37°C for 4 hours.
The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.3 ~g of a DNA fragment of about 0.41 kb was recovered. Next, a 3 ug portion of pAGE28 (Mizukami et al., J. Hiochem., 101, 1307-1310 (1987)) was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH 7.5), 7 mM magnesium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of ApaI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 2 ug of a DNA fragment of about 2.45 kb was recovered. Next, 0.1 ~g of the pKM641HA3 EcoRI-S, tvI fragment (about 0.41 kb) and 0.1 ~g of the pAGE28 EcoRI-ApaI fragment (about 2.45 kb) prepared above and 0.3 ~g ~~~1~~3~
of the synthetic DNA of SEQ ID N0:7 were dissolved in 20 ~tl of the T4 ligase buffer solution. The resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours . Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain a plasmid pKM641HE1 as shown in Fig. 24.
Since the thus constructed plasmid pKM641HE1 lacks a leader sequence, the following attempt was made to supplement the plasmid with the leader sequence using the synthetic DNA of SEQ ID NO: 6.
A 3 ~g portion of the plasmid pKM641HE1 was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HCl (pH
7.5), 7 mM magnesium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of ApaI and incubated at 37°C fox 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.4 ug of a DNA fragment of about 0.42 kb was recovered. Next, a 0.4 ~g portion of the thus prepared pKM641HE1 EcoRI-ApaI fragment (about 0.42 kb) was dissolved in 30 ul of a buffer solution containing ZO mM Tris-HC1 (pH 7.5), 7 mM magnesium chloride, 50 mM sodium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of Alul and incubated at 37 °C for 4 hours . The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation and about 0.3 ~g of a DNA fragment of about 0.4 kb was recovered.
_ sn _ Next, 0.1 ~g of the pKM641HE1 AluI-ApaI fragment (about 0.4 kb) and 0.1 ug of the pAGE28 EcoRI-ApaI fragment (about 2.45 kb) prepared above and 0.3 ug of the synthetic DNA of. SEQ
ID N0:6 were dissolved in 20 ~l of the T4 ligase buffer solution. The resulting solution was mixed with 350 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid -DNA, transformation of E.
coli HB101 was carried out to obtain a plasmid pKM641HF1 as shown in Fig. 25.
Next, immunoglobulin H chain~variable region of the thus obtained plasmid pKM641HF1 was introduced into the aforementioned cassette vector pChiIgHB2 in the following manner.
A 3 ~g portion of the plasmid pKM641HF1 was dissolved in 30 ul of a buffer solution containing 10 mM Tris-HC1 (pH
7.5), 7 mM magnesium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of A, naI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.5 ~g of a DNA fragment of about 0.44 kb was recovered. Next, a 3 ug portion of the pChiIgHB2 was dissolved in 30 ul of a buffer solution containing lO mM Tris-HC1 (pH
7.5), 7 mM magnesium chloride and 6 mM 2-mercaptoethanol. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of A~aI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to phenol--chloroform extraction _ 61 _ ~~l~~j and ethanol precipitation and about 3 ug of DNA was recovered.
Next, 0.1 ~g of the pKM641HF1 EcoRI-Apal fragment (about 0.44 kb) and 0.1 ug of the pChiTgHB2 EcoRI-ApaI fragment (about 10.1 kb) prepared above were dissolved in 20 ~1 of the T4 ligase buffer. The resulting solution was mixed with 350 units of T4 DNA'ligase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried out to obtain a plasmid pChi641HA1 as shown in Fig. 26.
Next, KM50~-derived immunoglobulin H chain promoter and enhancer regions of the thus obtained plasmid pChi641HA1 were converted into MoLTR in the following manner.
A 3 ~g portion of the plasmid pChi641HA1 was dissolved in 30 ~1 of a buffer solution containing 50 mM Tris-HC1 (pH
7.5), 10 mM magnesium chloride, 50 mM sodium chloride and 1 mM
DTT. The thus prepared solution was mixed with 10 units of EcoRI and 10 units of XhoI and incubated at 37°C for 4 hours.
The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.2 ~g of a DNA fragment of about 8.8 kb was recovered. Next, a 3 ~g portion of the pPMnL3 prepared in procedure 2 of Example 1 was dissolved in 30 dal of a buffer solution containing 50 mM Tris-HC1 (pH 7.5), 10 mM magnesium chloride, 50 mM sodium chloride and 1 mM DTT. The thus prepared solution was mixed with ZO units of EcoRI and 10 units of XhoI and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.3 ug of a DNA fragment of about 0.63 kb containing MoLTR was recovered. Next, 0.1 fag of the pChi641HA1 EcoRI-XhoI
fragment and 0.1 ug of the pPMOL3 EcoRI-XhoI fragment prepared above were dissolved in 20 ~l of the T4 ligase buffer solution.
The resulting solution was mixed with 175 units of T4 DNA
ligase and incubated at 4°C for 24 hours. Using the resulting reaction mixture, transformation of E. coli HB101 was carried out to obtain a plasmid pChi641HAM1 (Fig. 27) as a KM-641 chimera H chain expression vector.
6. Construction of KM-641 chimera L chain expression vector L chain variable region gene obtained by cleaving the plasmid pKM641LA2 variable region gene at its 5'-end EcoRI site and 3'-end ~IindIII site was ligated wi-ch the cassette vector for the expression of chimera L chain, using the synthetic DNA
shown in SEQ ID N0: 8, thereby constructing an L chain expression vector pchi641LG11 (Fig. 28).
Firstly, the DNA of SEQ ID N0: 8 (see Fig. 29) was synthesized using a DNA synthesizer. This synthetic DNA
comprises a base sequence corresponding to a region of the plasmid pKM641LA2 ranging from the 3' -end of the immunoglobulin L chain variable region to a HindIII cleavage site in the vicinity of the 3'-end and a base sequence corresponding to a region of the plasmid pChiIgLAl ranging from the 5'-end to an EcoRV cleavage site in the vicinity of the 5'-end. Thus, it has a HindIII cleavage site and an EcoRV cleavage site on both ends. Next, the thus synthesized DNA was introduced into the ~~ ~~~?~
plasmid pKM641LA2 in the following manner.
A 3 ug portion of the plasmid pKM641LA2 was dissolved in 30 ~1 of a buffer solution containing 10 mM Tris-HC1 (pH
7.5), 7 mM magnesium chloride, 50 mM sodium chloride and 6 mM
?.-mercaptoethanol. The thus prepared solution was mixed with units of EcoR2 and 10 units of HindII2 and incubated at 37 °C
for 4 hours. The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.3 ~g of a DNA fragment of aboutØ35 kb was recovered. Next, a 3 ug portion of pChiTgLAI was dissolved in 30 ~l of a buffer solution containing 50 mM Tris-HC1 (pH 7.5), 10 mM magnesium chloride, 50 mM sodium chloride and 1 mM DTT. The thus prepared solution was mixed with 10 units of EcoR2 and 10 units of EcoRV and incubated at 37°C for 4 hours. The resulting reaction mixture was subjected to phenol-chloroform extraction and ethanol precipitation and about 3 fag of DNA was recovered and dissolved in 10 ~1 of the TE solution (a buffer solution containing 10 mM
Tris-HC1 and 1 mM EDTA (pH 7.5)). Next, 0.1 ug of the pKM641LA2 EcoRT-HindTT2 fragment (about 0.35 kb) and 0.1 pg of the pChiIgLA1 EcoRT-EcoRV fragment (about 9.7 kb) prepared above and 0.3 ug of the s~rnthetic DNA of 5EQ ID N0:8 were dissolved in 20 ~1 of the T4 lipase buffer solution. The resulting solution was mixed with 350 units of T4 DNA lipase and incubated at 4°C for 24 hours. Using the thus obtained recombinant plasmid DNA, transformation of E. coli HB101 was carried oHt to obtain a plasmid pChi641LG11 as shown in Fig. 29.

~~"1~~
Next, KM50-derived immunoglobulin H chain promoter and enhancer regions of the thus obtained plasmid pChi641LG11 were converted into MoLTR in the following manner.
A 3 ug portion of the plasmid pChi641LG11 was dissolved in 30 ~1 of a buffer solution containing 50 mM Tris-I-IC1 (pH
7.5'), 10 mM magnesium chloride, 50 mM sodium chloride and 1 mM
DTT. The thus prepared solution was mixed with 10 units of EcoRT arid 10 units of XhoI and incubated at 37°C for 4 hours.
The resulting reaction mixture was subjected to agarose gel electrophoresis and about 0.2 ~g of a DNA fragment of about 8.3 kb was recovered. Next, 0.1 ~g of the pChi641hG11 EcoRI-XhoI
fragment and 0.1 ~.g of the pPMOL3 EcoRI-XhoI fragment prepared above were dissolved in 20 ~1 of the T4 ligase buffer. The resulting solution was mixed with 175 units of T4 DNA ligase and incubated at 4°C for 24 hours. Using the resulting reaction mixture, transformation of E. coli HB101 was carried out to obtain a plasmid pChi641LGM11 (Fig. 30) as a kcP2-641 chimera L chain expression vector.
7. Expression of anti-GD3 chimera antibody in SP2/0 cells Introduction of plasmid into SP2/0 cells taas carried out making use of the electroporation technique in accordance with the method of Miyaji et al. (Cytotechnology, 3, 133 - 140 (1990)).
The plasmids pChi641LG11 and pChi641HA1 (2 ~cg for each), or the plasmids pChi.641LGM11 and pChi641HAM1 (2 ~g for each), were simultaneously introduced into 2 x 106 of SP2/0 cells, and the resulting cells were suspended in 40 ml of RPMI1640-FCS(10) which has been prepared by supplementing RPMI1640 medium (Nissui Pharmaceutical Co., Ltd.) with 10~ of FCS, 1/40 volume of 7.5~ NaHC03, 3~ of 200 mM L-glutamine solution (available from GIBCO) and 0.5~ of a penicillin-streptomycin solution (GIBCO, a solution containing 5,000 units/ml of penicillin and 5,000 units/ml of streptomycin).
The thus prepared cell suspension was distributed in 200 ~.1-portions~into wells of a 96-well microtiter plate (Flow Laboratories ) , and the cells were cultured at 37 °C in a COZ
incubator. After 24 hours of the culturing, G41E (GIBCO) was added to the cell suspension to a final concentration of 0.5 mg/ml, and the culturing was continued for 1 to 2 weeks. When transformant colonies were developed and grown into confluent stages, culture broths were recovered from the wells to measure anti-GD3 chimera antibody activities by ELISA method in the following manner.
<Enzyme immunoassay (ELISA)> .
A 2 ng portion of GD3 (available from Iatron) or other type of ganglioside was dissolved in 2 ~1 of ethanol solution containing 5 ng of phosphatidylcholine ( Sigma Chemical Co . ) and 2.5 ng of cholesterol (Sigma Chemical Co.). A 20 ul portion of the thus prepared solution or the same volume of its dilution solution was distributed into each well of a '96-well microtiter plate (available from Greiner). After air-drying, blocking was effected with PBS containing 1~ BSA. To each well was added 50 ~;~~~~ 3;
to 100 ul of a culture supernatant of a transformant, a purified mouse monoclonal antibody solution or a purified chimera antibody solution. After reaction at 4°C for 10 hours, each well was washed with PBS and charged with 50 to 100 ~1 of peroxidase-labeled protein A (Funakoshi Pharmaceutical Co., Ltd'.), followed by 1 to 2 hours of reaction at room temperature. After washing with PBS, 50 to 100 ~1 of ABTS
substrate solution prepared by dissolving 550 mg of diammonium 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonate) in 0.1 M
citrate buffer (pH 4.2) and adding 1 ul/ml of hydrogen peroxide to the resulting solution just before its use was added to each well to develop color, and ODul3 of the reaction mixture was measured.
Among clones thus obtained, culture broth of a clone having the highest activity measured by ELTSA method contained anti-GD3 chimera antibody in an amount of about 0.1 ~g/ml.
The clone having anti-GD3 chimera antibody activity was suspended in the aforementioned RPMI1640-FCS(10) medium supplemented with 0.5 mg/ml of 6418 and 50 nM of methotrexate (to be referred to as "MTX" hereinafter) to a final cell density of 1-2 x 105 cells /ml. The thus prepared cell suspension was distributed in 2-ml portions into wells of a 24 well plate, and the cells were cultured at 37°C for 2 to 3 weeks in a COz incubator to induce clones resistant to 50 nM
MTX. When the thus induced clones were grown into confluent stages, anti-GD3 chimera antibody activities in the culture 67 _ broths were measured by the ELISA method. Among clones thus obtained, culture broth of a 50 nM MTX-resistant clone having the highest activity measured by ELISA method contained anti-GD3 chimera antibody in an amount of about 0.3 ~g/ml.
The 50 nM M'PX-resistant clone was suspended in the RPMI1640-FCS ( 10 ) medium supplemented with 0 . 5 mg/ml of 6418 and 200 nM of MTX to a final cell density of 1-2 x 105 cells /ml.
The thus prepared cell suspension was distributed in 2-ml portions~into wells of a 24 well plate, and the cells were cultured at 37°C for 2 to 3 weeks in a COZ incubator to induce clones resistant to 200 nM MTX. When the thus induced clones were grown into confluent stages, anti-GD3 chimera antibody activities in the culture broths were measured by the ELISA
method. Among clones thus obtained, culture broth of a 200 nM
MTX-resistant clone having the highest activity measured by ELISA method contained anti-GD3 chimera antibody in an amount of about 2 ~g/ml. This 200 nM MTX-resistant clone was named transformant KM-871.
Expression of anti-GD3 chimera antibody protein in the transformant KM-871 was confirmed by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) in the following manner.
The transformant KM-871 was suspended in GIT medium (Nihon Seiyaku Co., Ltd.) supplemented with 0.5 mg/ml of 6418 and 200 nl~i of MTX to a final cell density of 1-2 x 105 cells /ml. The thus prepared cell suspension was distributed in 100-ml portions in 175 cm2 flasks (available from Greiner), and the cells were cultured at 37°C for 3 to 5 days in a COZ incubator.
When the cells were grown into confluent stage, the resulting culture broth (about 900 ml) was recovered and subjected to salting-out with 50~ ammonium sulfate. Using Affigel'''H' Protein A MAPS-II Kit (Bio-Rad Laboratories), about 100 ~g of purified anti-GD3 chimera antibody KM-871 was obtained. About 5 ~g of the thus purified anti-GD3 chimera antibody KM-871 was subjected to electrophoresis in accordance with the conventional method (Laemmli, Nature, 227, 680 ( 1970 ) ) to check its molecular weight. The results are shown in Fig. 31. As shown in the figure, under reductive conditions, molecular weights of the chimera H chain and the chimera L chain were found to be about 50 kilodaltons and about 25 kilodaltons, respectively, thus confirming expression of the H and L chains having correct molecular weights. Under non-reductive conditions, molecular weight of the chimera antibody was found to be about 150 kilodaltons which confirmed expression of the correct size antibody consisting of two H chains and two L
chains.
8. Reaction specificity of anti-GD3 chimera antibody KM-871 Reactivities of the anti-GD3 chimera antibody with ganglioside GM1, N-acetyl GMZ (Boehringer-Mannheim Corp. ), N-glycolyl GMz, N-acetyl GM3, N-glycolyl GM3, GDIa, GDIb ( Iatron ) , GD2, GD3 ( Iatron ) , GTIb ( Funakoshi Pharmaceutical Co . , Ltd . ) and GQlb (Iatron) were measured by the ELISA method. In this instance, GM1 and GDIa were purified from bovine brain, N-~~~~eJ~
glycolyl GMz and N-glycolyl GM3 from mouse liver, N-acetyl GM3 from dog erythrocytes and GDZ from a cultured cell line IMR32 (ATCC CCL127), in accordance with the conventional method {J.
Biol. Chem., 263, 10915 (1988)). The results are shown in Table 1.
Table 1 Binding activity of antibody (OD~1~_!
Anti-GD3 chimera Mouse anti-GD3 Ganalioside antibody (0..3 ua/mll antibody (0.4 ua/m11.
N-acetyl GM3 0.007 0.006 N-glycolyl GM3 0 0 N-acetyl GMa 0 0 N-glycolyl GMz o 0 GD3 0.717 1.33 GDIa 0 0 GDIb 0 0 GTlb 0 ~ 0 GQIb p 0 .16 As shown in Table 1, anti-GD3 chimera antibody KM-8?1 and mouse anti-GD3 antibody KM-641 reacted only with GD3, thus showing no changes in the reaction specificity by the chimera formation.
9. Reactivity of anti-GD3 chimera antibody KM-871 by '~ n' 63 ~~~~~~n fluorescent antibody technique The cultured human malignant melanoma SK-MEZ-28 (ATCC
HTB72) and 6361 cells (;JCRB) both of which produced ganglioside GD3 were placed in a microtube (Treff ) to give a cell number of 1 x 106 cells per tube and washed by centrifugation (1,200 rpm, minutes) with PBS. 50 ~l of anti-GD3 chimera antibody KM-871 (10 ~g/ml) was added to the microtube and the mixture was allowed to react at 4°C for 30 minutes. Thereafter, the cells were washed three times by centrifugation (1,200 rpm, 5 minutes) with PBS, then 20 ul of fluorescein isocyanate-labeled Protein A (Boehringer Mannheim-Yamanouchi, 30-fold diluted) was added and, after stirring, the mixture was allowed to react at 4°C for 30 minutes. thereafter, the cells were washed three times by centrifugation (1,200 rpm, 5 minutes) with PBS, further suspended in PBS, and submitted for analysis using FCS-1 flow cell sorter (Nippon Bunko).
Control tests without the addition of KM-871 were performed by the same analytical procedure as mentioned above.
The results are shown in Fig. 32. The fluorescence intensity peak for KM-871 shaved shifting to the right (increased fluorescence intensity) as compared with the control, indicating that this antibody had reacted directly with ganglioside GD3 on the surface of the SK-MEL-28 and 6361 cells.
10. In vitro antitumor effect of anti-GD3 chimera antibody KM-871 (complement-dependent cytotoxicityeCDC) ~"l~~a~
(a) Preparation of target cells Suspensions of the target cells, namely SK-MEL-28 cells and 6361 cells, in RPMT-1640 medium supplemented with 10~ FCS
were respectively adjusted to a cell concentration of 1 x 10' cells/ml, NazsiCr04 was added to a concentration of 100 ~Ci/1 x 107'cells, reaction was performed at 37°C for 1 hour and, thereafter, 'the cells were washed three timES with the medium.
The cells were allowed to stand in the medium at 4°C for 30 minutes for spontaneous dissociation and then centrifuged (1,200 rpm, 5 minutes), and the medium was added to adjust the cell concentration to 4 x 106 cells/ml.
(b) Preparation of complement Serum from three healthy subjects were mixed to serve as a source of human complement.
(c) CDC activity measurement To U-bottomed 96-well plates was added anti-GD3 chimera antibody KM-871 or anti-GD3 mouse antibody KM-641 to final concentrations within the range of,0.05 ~g/m1 to 50 ~g/ml. To each well were added 2 x lOs target cells. Reaction was performed at room temperature for 1 hour. The supernatants were removed by centrifugation (1,200 rpm, 5 minutes), the complement solution prepared as described above under (b) was added in 150-ul portions (final concentration of 15 v/v~), and reaction was performed at 37°C for 1 hour. After centrifugation (1,200 rpm, 5 minutes), the amount of slCr in each supernatant was determined using a '~-counter. The amount of SiCr resulting from spontaneous dissociation was determined by adding to target cells the medium alone in place of the antibody and complement solution and determining the amount of S~Cr in the supernatant in the same manner as described above.
The total amount of free SzCr was determined by adding 5 N
sodium hydroxide in place of the antibody and complement solution, proceeding as described above, and determining the amount of SlCr in the supernatant.
The CDC activity was calculated as follows: ' CDC activity (~) _ .Amount of SICr resulting Amount of SICr in - from spontaneous sample supernatant dissociation x 100 Amount of Cr resulting Total amount of - from spontaneous free SlCr dissociation The results are shown in Fig.33. It was found from the results that chimera antibody KM-871 showed strong cytotoxicity against the SK-MEL-28 and 6361 cells as compared to mouse antibody KM-641, which indicates that chimera antibody KM-871 would be clinically more useful than mouse antibody KM-641.
11. In vitro antitumor effect of anti-GD3 chimera antibody KM-871 (antibody-dependent cell-mediated cytotoxicity:ADCC) (a) Preparation of target cells The target SK-MEL-28 and 6361 cells were prepared in the same manner as desdribed above under 10 (a).
(b) Preparation of effector cells 50 ml of human venous blood was collected, 0.5 ml of heparin sodium (Takeda Chemical Industries, 1,000 units/ml) was added, and the mixture was stirred gently and then centrifuged (1, 500 to 1, 800 g, 15 minutes) using PolymorphprepT°'' (Nycomed Pharma AS). The layer of lymphocytes and polymorphonuclear leukocytes was separated, and the cells were washed three times by centrifugation ( 1, 500 to 1, 800 g, 15 minutes ) with PRMI-1640 medium and suspended in RPMI-1640 medium supplemented with 10~
FCS (5 x106 cells/ml) for use as effector cells.
(c) ADCC activity measurement To U-bottomed 96-well plates was added anti-GD3 chimera antibody KM-871 or anti-GD3 mouse antibody KM-641 in 50-ul portions to final concentrations of 10 ~g/ml. To each well were added 100 ul of target cells (2 x 105 cells) and 50 ~l of effector cells (5 x 105 cells) so that the ratio of effector cells to target cells should be 50:1 or 100:1. Reaction was performed at 37°C for 4 hours, followed by centrifugation (1,200 rpm, 5 minutes). The amount of SICr in each supernatant was determined using a '~-counter. The amount of 5'Cr resulting from spontaneous dissociation was determined by adding to target cells the medium alone in place of the antibody and effector cells and measuring the amount of 5lCr in the supernatant in the same manner as described above. The total amount of free SICr was determined by adding S N sodium hydroxide in place of the antibody and effector cells, proceeding as described above, and determining the amount of ~~~1~
siCr in the supernatant.
The ADCC activity was calculated as fol7.ows:
ADCC activity (~) _ .Amount of slCr Amount of slCr in - resulting from sample supernatant spontaneous dissociation x 100 Amount of Cr Total amount of - resulting from free s'Cr spontaneous dissociation As a control, the medium was added in place of the antibodies, the procudure mentioned above was followed, and the amount of SICr in 'the control supernatant was determined for ADCC activity calculation.
The results are shown in Fig. 34. Tn both cases of using lymphocytes and polymorphonuclear leukocytes as effector cells, chimera antibody KM-871 showed strong antibody-dependent cell-mediated cytotoxicity against the 6361 cells as compared to mouse antibody KM-641, which indicates that chimera antibody KM-871 would be clinically more useful than mouse antibody KM--641.
12. In vivo therapeutic effect of anti-GD3 chimera antibody KM-871 (therapeutic effect on transplanted tumors) Human malignant melanoma 6361 cells (7. x 10' cells) were intracutaneously transplanted to abdominal paxts of Balb/c nu/nu mice (5 to 7 aminals/group). Anti-GD3 chimera antibody KM-871 (100 ~g/animal) was intravenously administered into mice _ 75 _ ~~"l!~
four times starting from the next day of the transplantation of the tumor cells. To the mice of the control group, 100 ~g of anti-GD3 mouse antibody KM-641 or anti-Sialyl Lea monoclonal antibody AMC-462 (ECACC 86050801) was intravenously administered five times starting from the day of the trai~splan~tation. The therapeutic effect on transplanted tumor cells was determined in terms of tumor size (volume) calculated by the following equation.
Tumor size (mm3) = 0.4 x a x bz a: major axis b: minor axis The results are shown in Fig. 35. As shown in Fig. 35, remarkable growth of tumors was observed in the control group to which AMC-462 was administered, while the growth of tumors was significantly suppressed in the group to which KM-641 was administered. KM-871 showed further stronger therapeutic effect so that the establishment of tumors was completely inhibited 65 days after the transplantation.
Reference Example.l (1) Preparation of antigen In 30 ml of chloroform/methanol (2/1) solution were dissolved 5 ~g of ganglioside GD3 having NeuAca2-~BNeriAca2-a3Ga1 sugar chain on its non-reducing end (Iatron), 0.5 umol of dipalmitoylphosphatidylcholine (Sigma Chemical Co.), 0.5 umol of cholesterol (Nakalai Tesque), 0.05 umol of dipalmitoylphosphatidylic acid (Sigma Chemical Co.) and 2.5 ~.g of Lipid A (Funakoshi Pharmaceutical Co., Ltd.). The thus prepared solution was warmed at 45°C to remove solvents, thereby obtaining a uniform lipid thin film. After completely removing solvents by sacking the film for 1 hour using a vacuum pump, the resulting film was mixed with 0.5 ml of PBS solution and stirred at 45°C to obtain an antigen solution.
(2) Preparation of antibody-producing cells Mice was immunized by administering 0.5 ml of the antigen solution obtained in the above step ( 1 ) into the caudal vein once every week for 7 weeks. For further immunization, ganglioside GD3-positive SiC-MEL-28 (ATCC HTB 72 ) cells ( 1 x 10' cells) were intraperitoneally administered once every week for three weeks. On the third day after the last administration,' spleen cells were prepared from each mouse for use in the following cell fusion.
(3) Preparation of mouse myeloma cells A mouse myeloma cell line P3-U1 having 8-azaguanine resistance was cultured in normal medium (RPMI1640 medium containing 10~ fetal calf serum (FCS) ) to obtain 2 x 10' or more cells for use in the following cell fusion as parent cells.
(4) Preparation of hybridoma The spleen cells and myeloma cells obtained in the above steps (2) and (3), respectively, were used in ratio of r,s ~.~ >.~ c~ rl 10:1 and subjected to cell fusion in accordance with the aforementioned procedure. After culturing at 37°C for 14 days in HAT medium (prepared by supplementing normal medium with hypoxanthin (10'4 M), thymidine (1.5 x 10'5 M) and aminopterine ( 4 x 10'' M) ) under an atmosphere of 5~ CO2, fused cells were selected and cultured in FIT medium (HAT medium minus aminopterine). Then, active wells were selected by assaying the antibody titers against ganglioside GD3, and after changing to normal medium, cloning was repeated twice. Thereafter, hybridomas which showed specific reaction with ganglioside GD3 were selected by enzyme immunoassay or immunohistological evaluation (ABC method). That is, 2 ng of ganglioside GM3 (purified from dog erythrocytes in accordance with the method of Nores et a1. , J. Immunol . , 139, 3171 ( 1987 ) ) and 2 ng of ganglioside GD3 (Iatron) were dissolved in 2 ml of ethanol solution containing 5 ng of phosphatidylcholine (Sigma Chemical Co.) and 2.5 ng of cholesterol (Sigma Chemical Co.). The thus prepared solution was distributed in 20-~.1 portions into wells of a 96 well microtiter plate (Flow Laboratories), air-dried and then subjected to blocking using 1~ BSA-PBS solution. Each of the resulting hybridoma culture supernatant was distributed in 50-~1 portions into the plate wells carrying a ganglioside GD3 adsorbed and the plate carrying ganglioside GM3 adsorbed thereon, and the reaction was allowed to proceed at 4°C for 18 hours.
After the reaction, a hybridoma strain capable of .- 78 _ producing mouse monoclonal antibody specifically reactive with ganglioside GD3 but not with ganglioside GM3 were selected in accordance with the known method (Cancer Res., 46, 4438 (1986)). This mouse monoclonal antibody, was named "mouse monoclonal antibody ICM-641", and the hybridoma which produces this antibody was named "hybridoma KM-641". The hybridoma KM-641 has been deposited on September 27, 1990, with Fermentation ' Research Institute, Agency of Industrial Science and Technology, under the Budapest Treaty and has been assigned the designation as FERM BP-3116.
The present invention provides a process for the production of humanized chimera antibody wherein the chimera antibody can be produced easily without changing any one of amino acids of its mouse antibody variable region, as well as a humanized chimera antibody specific for ganglioside GD3.
While the invention has been described in detail and with reference to specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope of the invention.
_ 79 _ r r~ P
~1%~
SEQUENCE LISTING
SEQ ID N0:1:

SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 812 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: DNA (genomic) ORIGINAL SOURCE:

IMMEDIATE EXPERIMENTAL SOURCE

NAME OF CELL LINE: hybridoma FEATURES:

from 256 to 262 by TATA signal TTC CTG

Met Asp Arg Leu Thr Ser Ser Leu Leu Met Val Pro Ala Phe Leu GTGAGTACCA .AAGCTTCCTA AGTGATGAACTGTTCTATCCTCACCTGTTC AAACCTGACC405 AT GT CAG GTT
ACT CTG
AAA

Tyr Va l Leu Ser Gln Val Thr Leu Lys _4 1 5 CAG CAG

Glu Ser Gly Pro G1y Ile Leu Pro Ser Thr Leu Ser Leu Thr Gln Gln CTG TAT

Cys Ser Phe Ser Gly Phe Ser Ser Thr Gly Met Cys Val Gly Leu Tyr AAG GAG

Trp Ile Arg Gln Ser Ser Gly Gly Leu Trp Leu Ala Asn Val Lys Glu TAC TCT

~~'~1~
Trp Trp Ser Ala Tyr Asn ProSer Leu LysAsn ArgLeu Asp Lys Tyr AAG ACC AAC

Thr Ile Ser Asp Ser Asn GlnAla Phe LeuLys IleThr Lys Thr Asn ACT GAT GCC

Asn Met Asp Ala Thr I1e TyrTyr Cys AlaGly ArgGly Thr Asp Ala GGT GTG TTT

Ala Thr Glu Ile Sex Asp TyrTrp Gly HisGly ValMet Gly Val Phe TCC GGTAAG

Val Thr Val Ser Ser 120 ' SEQ ID N0:2:

SEQUENCE TYPE:46 basepairs SEQUENCE LENGTH: nucleic acid STRANDEDNESS:ouble d TOPOLOGY: r linea MOLECULE TYPE:other cleic id, synthetic nu ac DNA

GGGC TCAAGCTTGT
CGACTC

SEQ ID N0:3:
SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 29 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: other nucleic acid, synthetic DNA

SEQ ID N0:4:
SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 403 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: cDNA to mRNA
ORIGINAL SOURCE:

IMMEDIATE
EXPERIMENTAL
SOURCE

NAME OF CELLLINE: hybridomaKM-641 FEATURES:

From 14 to 3 sig peptide IDENTIFICATI ON METHOD:
S

G CTT GTT TTC GGT
A GTT
CAG
TGT
GAA
GTG

Leu Val Leu.Val Phe Lys Gly Val Gln Cys Glu Val _10 _5 1 ACG CTG GAG TCT GGG GACTTT GTGAAA CCTGGA.GGGTCC CTG 97 GTG GGA

Thr Leu Glu Ser Gly AspPhe ValLys ProGly GlySer Leu Val Gly TCC TCT

Lys Val Cys Ala Ala GlyPhe AlaPhe SerHis TyrAla Met Ser Ser GTT CCG

Ser Trp Arg Gln Thr AlaLys ArgLeu GluTrp ValAla Tyr Val Pro AAG

IleSer SerGly GlySer GlyThr TyrTyr Ser AspSer ValLys Gly ArgPhe ThrIle SerArg AspAsn AlaLys Asn ThrLeu TyrLeu Gln MetArg SerLeu ArgSer GluAsp SerAla Met TyrPhe CysThr Arg GTTAAA CTGGGA ACGTAC TACTTT GACTCC TGG GGCCAA GGCACC ~ACT 385 ValLys LeuGly ThrTyr TyrPhe AspSer Trp GlyGln GlyThr Thr Leu Thr Val Ser Ser Ala SEQ ID N0:5:
SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 408 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: cDNA to mRNA
ORIGINAL SOURCE:
IMMEDIATE EXPERIMENTAL SOURCE
NAME OF CELL LINE: hybridoma KM-641 FEATURES:
from 25 to 84 sig peptide IDENTIFICATION METHOD: S

GAGTCAGCCT
GGAC
ATG
ATG
TCC
TCT
GCT
CAC
TTC
CTT
GGT

Met Met Ser Ser Ala Gln Phe Leu Gly - 2p -15 LeuLeu Leu LeuCys PheGln GlyThr ArgCys Asp IleGln MetThr GlnThr Ala SerSer LeuPro AlaSer LeuGly Asp ArgVal ThrIle SerCys Ser AlaSer GlnAsp IleSer AsnTyr Leu AsnTrp Tyr.Gln GlnLys Pro AspGly ThrVal LysLeu LeuIle Phe TyrSer SerAsn LeuHis Ser GlyVal ProSer ArgPhe SerGly Gly GlySer GlyThr AspTyr Ser LeuThr I12Sex AsnLeu GluPro Glu AspI12 AlaThr Tyr Phe Cys His Gln Tyr Ser Lys Leu Pr_o Trp Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg SEQ ID N0:6:
SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 35 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: other nucleic acid, synthetic DNA

Met Glu Phe Gly Leu Ser Trp Leu Phe SEQ ID N0:7:
SEQUENCE TYPE: nucleic acid SEQUENCE LENGTH: 43 base pairs STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: other nucleic acid, synthetic DNA

ACT TCC' ACC

Gln Gly Thr Thr Leu Val Ser AlaSer Lys Gly Thr Ser Thr SEQ ID N0:$:

SEQUENCE TYPE: nucleicacid SEQUENCE LENGTH: e pairs 61 bas STRANDEDNESS: double TOPOLOGY: linear MOLECULE TYPE: othercleic synthetic nu acid, DNA

TTC GGA CTG

Leu Pro Trp Thr Phe Gly Gly ThrLys Glu Ile Lys Arg Gly Leu Thr Val Ala Ala Leu Val Leu Val Phe Lys G1y Val Gln Cys Glu Val Thr Leu Val Glu Ser Gly Gl.y Asp Phe Val Lys Pzo GIy Gly Ser Leu Lys Val Ser Cys Ala Ala Ser Gly Phe Ala Phe S~:r His Tyr Ala Met Ser Trp Val Arg Gln Thr Pro Ala Lys Arg Leu Glu Trp Val A.la Tyr Ile Ser Ser Gl.y Gly Ser Gly Thz- Tyr Tyr 5er Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Leu Tyr Leu G1n Met Arg Scr Leu Arg Ser Glu Asp Ser Ala Met Tyr Phe Cys Thr Arg Val L,ys Leu Gly Thr Tyr Tyr Phe Asp Ser Trp Gly Gln Gly Thr Thr Leu Thr Val. Ser Ser Ala Met ~Iet Ser Ser Ala Gln Phe Leu G.l,y L eu Leu Lett Leu Cys 1'he Gln Gly Th.r Arg Cys Asp Ile Gln Met Thr Gln Thr Ala Ser Ser Leu Pro Ala Ser Leu Gly Asp Arg Val Thr I1e Sex Cys Ser Ala Ser Gln Asp Ile Ser Asn Tyr Leu Asn 'frp Tyr GIn Gln Lys Pro Asp Gly Thr Val Lys Leu Leu Tle Phe Tyr Ser Ser Asn Leu H1s Set Gly Val Pro Ser Ark Phe Ser Gly Gly Gly Ser Gly Thr Asp Tyr Sex Leu Thr Ile Ser $5 90 95 Asn Leu Glu Pro Glu Asp Ilc Ala Thr Tyr Phe Cys His Gln Tyr Ser Lys Leu Pro Trp Thr Sex Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg

Claims (29)

1. A process for producing chimeric antibody which comprises the steps of:
(1) constructing a cassette vector by inserting a cDNA coding for a heavy chain constant region of human antibody into an expression vector for animal cell use and establishing a cloning site in the upstream region of the heavy chain constant region of said cassette vector for inserting a cDNA which encodes a heavy chain variable region of nonhuman animal antibody;
(2) digesting a cDNA coding for the heavy chain variable region of nonhuman animal antibody with restriction enzymes;
(3) inserting said cDNA coding for the heavy chain variable region of nonhuman animal antibody into the cassette vector, using a synthetic DNA which comprises a nucleotide sequence corresponding to the 5'-end side of said heavy chain constant region of human antibody as defined by the 31st to 43rd nucleotide in SEQ ID NO: 7 and a nucleotide sequence corresponding to the 3'-end side of said heavy chain variable region of nonhuman animal antibody and is possessed of the restriction enzyme recognition sites on both of its ends, thereby constructing a chimeric antibody heavy chain expression vector in which said cDNA coding for the heavy chain constant region of human antibody and said cDNA coding for the heavy chain variable region of nonhuman animal antibody are linked together through said synthetic DNA;
(4) constructing a cassette vector by inserting a cDNA coding for a light chain constant region of human antibody into an expression vector for animal cell use and establishing a cloning site in the upstream region of the light chain constant region of said cassette vector for inserting a cDNA which encodes a light chain variable region of nonhuman animal antibody;

(5) digesting a cDNA coding for the light chain variable region of nonhuman animal antibody with restriction enzymes;
(6) inserting said cDNA coding for a light chain variable region of nonhuman animal antibody into the cassette vector, using a synthetic DNA which comprises a nucleotide sequence corresponding to the 5'-end side of said light chain constant region of human antibody as defined by the 48th to 61st nucleotide in SEQ ID NO: 8 and a nucleotide sequence corresponding to the 3'-end side of said light chain variable region of nonhuman animal antibody and is possessed of the restriction enzyme recognition sites on both of its ends, thereby constructing a chimeric antibody light chain expression vector in which said cDNA coding for the light chain constant region of human antibody and said cDNA coding for the light chain variable region of nonhuman animal antibody are linked together through said synthetic DNA;
(7) introducing these expression vectors into host cells to obtain a transformant; and (8) culturing said transformant in an appropriate culture medium, thereby allowing the transformant to produce and accumulate a chimeric antibody, and collecting said chimeric antibody from the resulting culture broth.
2. The process according to claim 1, wherein said nonhuman animal is mouse.
3. The process according to claim 1, wherein said nonhuman animal antibody is reactive with ganglioside.
4. The process according to claim 1, wherein said nonhuman animal antibody is reactive with ganglioside GD3.
5. The process according to claim 1, wherein said nonhuman animal antibody is reactive with mouse ganglioside GD3.
6. The process according to claim 1, wherein said heavy chain variable region of antibody comprises the polypeptide having the amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9 and said light chain variable region of antibody comprises the polypeptide having the amino acid sequence of residues 21 to 127 defined in SEQ ID NO: 10.
7. The process according to claim 1, wherein said chimeric antibody is reactive with ganglioside GD3.
8. The process according to claim 1, wherein said chimeric antibody is an antibody KM-871 produced by a hybridoma KM-871 (FERM BP-3512).
9. A chimeric antibody which comprises a variable region of a mouse monoclonal antibody KM-641 produced by a hybridoma KM-641 (FERM BP-3116) which is reactive with ganglioside GD3 and 3',8'-LD1 and a human antibody constant region.
10. A chimeric antibody KM-871 produced by a transformant KM-871 (FERM BP-3512) which is reactive with ganglioside GD3.
11. A transformant capable of producing a chimeric antibody, wherein said antibody comprises a variable region of a mouse monoclonal antibody KM-641 produced by a hybridoma KM-641 (FERM BP-3116) which is reactive with ganglioside GD3 and 3',8'-LD1 and a human antibody constant region.
12. A transformant KM-871 (FERM BP-3512) capable of producing chimeric antibody KM-871, wherein said antibody is reactive with ganglioside GD3.
13. A DNA which has the nucleotide sequence defined in SEQ ID NO: 7 or a linker DNA containing said DNA.
14. A DNA which has the nucleotide sequence defined in SEQ ID NO: 8 or a linker DNA containing said DNA.
15. A pharmaceutical composition comprising a chimeric antibody KM-871 produced by a transformant KM-871 (FERM BP-3512) which is reactive with ganglioside GD3 and a pharmaceutically acceptable carrier.
16. The use of a pharmaceutically acceptable amount of a chimeric antibody produced by a transformant KM-871 which is reactive with ganglioside GD3 to treat cancer.
17. A chimeric antibody comprising a heavy chain variable region and a light chain variable region of a nonhuman antibody, and a heavy chain constant region and a light chain constant region of a human antibody, wherein said chimeric antibody binds the ganglioside GD3 and said nonhuman antibody is a mouse monoclonal antibody KM-641 produced by a hybridoma KM-641 (FERM BP-3116).
18. The chimeric antibody according to claim 17, wherein said heavy chain variable region has the amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9.
19. The chimeric antibody according to claim 17, wherein said light chain variable region has the amino acid sequence of residues 21 to 127 defined in SEQ ID NO: 10.
20. A chimeric antibody comprising a heavy chain variable region and a light chain variable region of a nonhuman antibody, and a heavy chain constant region and a light chain constant region of a human antibody, wherein said chimeric antibody binds the ganglioside GD3, said heavy chain variable region has the amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9, and said light chain variable region has the amino acid sequence of residues 21 to 127 defined in SEQ ID NO: 10.
21. A polypeptide comprising an amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9.
22. A polypeptide comprising an amino acid sequence of residues 21 to 127 defined in SEQ ID NO: 10.
23. A DNA encoding an amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9 or residues 21 to 127 defined in SEQ ID NO: 10.
24. The DNA according to claim 23, comprising a nucleotide sequence of residues 44 to 400 defined in SEQ ID
NO: 4.
25. The DNA according to claim 23, comprising a nucleotide sequence of residues 85 to 405 defined in SEQ ID
NO: 5.
26. A DNA encoding a chimeric antibody comprising a heavy chain variable region and a light chain variable region of a nonhuman antibody, and a heavy chain constant region and a light chain constant region of a human antibody, wherein said chimeric antibody binds the ganglioside GD3, said heavy chain variable region has the amino acid sequence of residues 11 to 129 defined in SEQ ID NO: 9, and said light chain variable region has the amino acid sequence of residues 21 to 127 defined in SEQ ID NO: 10.
27. An expression vector comprising the DNA
described in any one of claims 23 to 26.
28. An isolated cell comprising the expression vector according to claim 27.
29. The cell according to claim 28, wherein said cell is an animal cell.
CA002078539A 1991-09-18 1992-09-17 Process for producing humanized chimera antibody Expired - Lifetime CA2078539C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP23837591 1991-09-18
JP3-238375 1991-09-18

Publications (2)

Publication Number Publication Date
CA2078539A1 CA2078539A1 (en) 1993-03-19
CA2078539C true CA2078539C (en) 2005-08-02

Family

ID=17029253

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002078539A Expired - Lifetime CA2078539C (en) 1991-09-18 1992-09-17 Process for producing humanized chimera antibody

Country Status (10)

Country Link
US (10) US5750078A (en)
EP (2) EP0533199B1 (en)
JP (1) JP3440104B2 (en)
AT (2) ATE239078T1 (en)
AU (2) AU669124B2 (en)
CA (1) CA2078539C (en)
DE (2) DE69233027T2 (en)
DK (1) DK0533199T3 (en)
ES (1) ES2195995T3 (en)
PT (1) PT533199E (en)

Families Citing this family (265)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU669124B2 (en) * 1991-09-18 1996-05-30 Kyowa Hakko Kirin Co., Ltd. Process for producing humanized chimera antibody
US5830470A (en) * 1992-09-07 1998-11-03 Kyowa Hakko Kogyo Co., Ltd. Humanized antibodies to ganglioside GM2
US6042828A (en) 1992-09-07 2000-03-28 Kyowa Hakko Kogyo Co., Ltd. Humanized antibodies to ganglioside GM2
US5939532A (en) * 1993-09-07 1999-08-17 Kyowa Hakko Kogyo Co., Ltd Humanized antibodies to ganglioside GM2
US6824777B1 (en) * 1992-10-09 2004-11-30 Licentia Ltd. Flt4 (VEGFR-3) as a target for tumor imaging and anti-tumor therapy
US7105159B1 (en) 1992-11-05 2006-09-12 Sloan-Kettering Institute For Cancer Research Antibodies to prostate-specific membrane antigen
EP0729976A1 (en) * 1993-11-19 1996-09-04 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human medulloblastomatous cell
US6088323A (en) * 1996-07-16 2000-07-11 Sony Corporation Optical disk, optical disk device, and optical disk recording method
US6986890B1 (en) 1996-11-21 2006-01-17 Kyowa Hakko Kogyo Co., Ltd. Anti-human VEGF receptor Flt-1 monoclonal antibody
WO1998022616A1 (en) 1996-11-21 1998-05-28 Kyowa Hakko Kogyo Co., Ltd. ANTI-HUMAN VEGF RECEPTOR F1t-1 MONOCLONAL ANTIBODY
US20030170248A1 (en) * 1997-12-23 2003-09-11 Jeffrey R. Stinson Humanized monoclonal antibodies that protect against shiga toxin induced disease
DE69940733D1 (en) * 1998-05-20 2009-05-28 Kyowa Hakko Kogyo Kk GEN RECOMBINANT ANTIBODIES
ATE276763T1 (en) 1998-07-13 2004-10-15 Univ Texas USE OF ANTIBODIES TO AMINOPHOSPHOLIPIDES FOR CANCER TREATMENT
CN100340291C (en) * 1998-10-09 2007-10-03 路德维格癌症研究院 F1t4(VEGFR-3) as target for tumor imaging and anti-tumor therapy
US7026449B2 (en) * 1999-01-05 2006-04-11 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP2270148A3 (en) 1999-04-09 2011-06-08 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
EP1238985B1 (en) * 1999-09-30 2008-03-05 Kyowa Hakko Kogyo Co., Ltd. Human type complementarity determining region transplantation antibody against ganglioside gd3 and derivatives of antibody against ganglioside gd3
EP1881004B1 (en) 2000-04-21 2012-07-25 Fuso Pharmaceutical Industries, Ltd. Novel collectin
DK1279677T3 (en) 2000-04-25 2007-05-07 Otsuka Pharma Co Ltd GD3 mimetic peptides
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
ES2639222T5 (en) 2000-10-06 2023-11-24 Kyowa Kirin Co Ltd Cells that produce antibody compositions
JP4669984B2 (en) 2001-01-19 2011-04-13 ベジェニクス リミテッド F1t4 (VEGFR-3) and antitumor therapy as a target for tumor imaging
EP1384487A4 (en) * 2001-03-29 2007-07-11 Kyowa Hakko Kogyo Kk Drugs containing genetically modified antibody against ganglioside gd3
US6820011B2 (en) * 2001-04-11 2004-11-16 The Regents Of The University Of Colorado Three-dimensional structure of complement receptor type 2 and uses thereof
WO2003024191A2 (en) * 2001-09-21 2003-03-27 Raven Biotechnologies, Inc. Antibodies that bind to cancer-associated antigen cytokeratin 8 and methods of use thereof
JP4347694B2 (en) * 2001-10-16 2009-10-21 レイベン バイオテクノロジーズ,インコーポレイティド Antibody binding to cancer-associated antigen CD46 and method of use thereof
US20110045005A1 (en) 2001-10-19 2011-02-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic origin
US20050215472A1 (en) * 2001-10-23 2005-09-29 Psma Development Company, Llc PSMA formulations and uses thereof
EP2360169B1 (en) * 2001-10-23 2015-10-14 Psma Development Company, L.L.C. PSMA antibodies
WO2003084569A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Drug containing antibody composition
AU2003230929A1 (en) * 2002-04-12 2003-10-27 Raven Biotechnologies, Inc. Antibodies that bind to integrin alpha-v-beta-6 and methods of use thereof
WO2003093443A2 (en) * 2002-05-03 2003-11-13 Raven Biotechnologies, Inc. Alcam and alcam modulators
US7893218B2 (en) 2003-06-16 2011-02-22 Stowers Institute For Medical Research Antibodies that specifically bind SOST peptides
PT1565489E (en) 2002-06-19 2011-02-23 Raven Biotechnologies Inc Internalizing antibodies specific for the raag10 cell surface target
ATE491725T1 (en) 2002-07-15 2011-01-15 Univ Texas ANTIBODIES BOUND TO ANIONIC PHOSPHOLIPIDES AND AMINOPHOSPHOLIPIDES AND THEIR USE IN THE TREATMENT OF VIRUS INFECTIONS
EP1545615A4 (en) * 2002-10-04 2006-03-01 Rinat Neuroscience Corp Methods for treating cardiac arrhythmia and preventing death due to cardiac arrhythmia using ngf antagonists
UA80447C2 (en) 2002-10-08 2007-09-25 Methods for treating pain by administering nerve growth factor antagonist and opioid analgesic
DK1556083T3 (en) * 2002-10-08 2011-04-04 Rinat Neuroscience Corp Method of treating post-operative pain by administration of an antibody to nerve growth factor and composition containing the same
US7255860B2 (en) * 2002-10-08 2007-08-14 Rinat Neuroscience Corp. Methods for treating post-surgical pain by administering an anti-nerve growth factor antagonist antibody
KR20050071564A (en) * 2002-10-09 2005-07-07 리나트 뉴로사이언스 코퍼레이션 Methods of treating alzheimer's disease using antibodies directed against amyloid beta peptide and compositions thereof
AU2003287622A1 (en) * 2002-11-06 2004-06-03 Fraunhofer Usa Expression of foreign sequences in plants using trans-activation system
US7692063B2 (en) * 2002-11-12 2010-04-06 Ibio, Inc. Production of foreign nucleic acids and polypeptides in sprout systems
US7683238B2 (en) * 2002-11-12 2010-03-23 iBio, Inc. and Fraunhofer USA, Inc. Production of pharmaceutically active proteins in sprouted seedlings
US7405061B2 (en) * 2002-11-13 2008-07-29 Raven Biotechnologies, Inc. Antigen PIPA and antibodies that bind thereto
US7276589B2 (en) * 2002-11-26 2007-10-02 Pdl Biopharma, Inc. Chimeric and humanized antibodies to α5β1 integrin that modulate angiogenesis
AU2003298783B2 (en) 2002-11-26 2010-11-04 Abbvie Biotherapeutics Inc. Chimeric and humanized antibodies to alpha5beta1 integrin that modulate angiogenesis
US7285268B2 (en) 2002-11-26 2007-10-23 Pdl Biopharma, Inc. Chimeric and humanized antibodies to α5β1 integrin that modulate angiogenesis
AU2003300397A1 (en) * 2002-12-23 2004-07-22 Rinat Neuroscience Corp. Methods for treating taxol-induced sensory neuropathy
CA2511598C (en) 2002-12-24 2016-09-13 Rinat Neuroscience Corp. Anti-ngf antibodies and methods using same
US7569364B2 (en) 2002-12-24 2009-08-04 Pfizer Inc. Anti-NGF antibodies and methods using same
US9498530B2 (en) 2002-12-24 2016-11-22 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
WO2004070016A2 (en) 2003-02-03 2004-08-19 Fraunhofer Usa Inc. System for expression of genes in plants
CA2516454A1 (en) 2003-02-19 2004-09-02 Rinat Neuroscience Corp. Methods for treating pain by administering a nerve growth factor antagonist and an nsaid and compositions containing the same
WO2004084836A2 (en) * 2003-03-20 2004-10-07 Rinat Neuroscience Corp. Methods for treating taxol-induced gut disorder
JP2007524605A (en) * 2003-04-03 2007-08-30 ピーディーエル バイオファーマ,インコーポレイティド Inhibitors of integrin α5β1 and their use for control of tissue granulation
CU23403A1 (en) * 2003-04-23 2009-08-04 Centro Inmunologia Molecular RECOMBINANT ANTIBODIES AND FRAGMENTS RECOGNIZING GANGLIOSIDE N-GLICOLIL GM3 AND ITS USE FOR DIAGNOSIS AND TUMOR TREATMENT
WO2005026375A2 (en) 2003-05-22 2005-03-24 Fraunhofer Usa, Inc. Recombinant carrier molecule for expression, delivery and purification of target polypeptides
US20050232926A1 (en) * 2003-06-06 2005-10-20 Oncomax Acquisition Corp. Antibodies specific for cancer associated antigen SM5-1 and uses thereof
CN1279056C (en) * 2003-06-06 2006-10-11 马菁 Specific antibody of tumor-associated antigen SM5-1 and use thereof
US20050131219A1 (en) * 2003-08-18 2005-06-16 Urdea Michael S. Methods for reducing complexity of a sample using small epitope antibodies
AU2004274487B2 (en) * 2003-09-18 2011-09-01 Macrogenics West, Inc. KID3 and KID3 antibodies that bind thereto
EP3006463A1 (en) 2003-10-01 2016-04-13 Kyowa Hakko Kirin Co., Ltd. Method for stabilizing antibody and stabilized solution-type antibody preparation
EP1700121A4 (en) 2003-12-23 2008-09-03 Rinat Neuroscience Corp Agonist anti-trkc antibodies and methods using same
EP1769068B1 (en) * 2004-02-20 2014-12-31 iBio, Inc. Systems and methods for clonal expression in plants
ATE531388T1 (en) * 2004-03-24 2011-11-15 Abbott Biotherapeutics Corp USE OF ANTI-ALPHA5BETA1 ANTIBODIES TO INHIBIT CANCER CELL PROLIFERATION
ES2338344T3 (en) * 2004-04-07 2010-05-06 Rinat Neuroscience Corporation PROCESS OF BONE CANCER TREATMENT THROUGH THE ADMINISTRATION OF AN ANTAGONIST OF THE NEURONAL GROWTH FACTOR.
CA2569692C (en) * 2004-06-07 2015-07-21 Raven Biotechnologies, Inc. Transferrin receptor antibodies
EP2495252B1 (en) 2004-07-09 2018-04-18 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Soluble forms of hendra and nipah virus G glycoprotein
US7927594B2 (en) 2004-07-30 2011-04-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide
CA2593786A1 (en) 2005-01-12 2006-07-20 Raven Biotechnologies, Inc. Kid31 and antibodies that bind thereto
EP1846032A4 (en) * 2005-01-31 2009-01-28 Raven Biotechnologies Inc Luca2 and antibodies that bind thereto
WO2006084078A2 (en) * 2005-02-02 2006-08-10 Raven Biotechnologies, Inc. Jam-3 and antibodies that bind thereto
AU2006210589B2 (en) * 2005-02-02 2011-12-08 Macrogenics West, Inc. ADAM-9 modulators
US7572896B2 (en) 2005-02-03 2009-08-11 Raven Biotechnologies, Inc. Antibodies to oncostatin M receptor
US7569672B2 (en) * 2005-02-04 2009-08-04 Raven Biotechnologies, Inc. Antibodies that bind to EphA2 and methods of use thereof
US20070003557A1 (en) * 2005-04-21 2007-01-04 Andres Forero Method for treating cancer using premedication
AR054260A1 (en) * 2005-04-26 2007-06-13 Rinat Neuroscience Corp METHODS OF TREATMENT OF DISEASES OF THE LOWER MOTOR NEURONE AND COMPOSITIONS USED IN THE SAME
UY29504A1 (en) * 2005-04-29 2006-10-31 Rinat Neuroscience Corp DIRECTED ANTIBODIES AGAINST BETA AMYLOID PEPTIDE AND METHODS USING THE SAME.
CN101282994B (en) 2005-07-22 2013-09-18 Y's治疗有限公司 Anti-CD26 antibodies and methods of use thereof
WO2007117264A2 (en) * 2005-08-03 2007-10-18 Fraunhofer Usa, Inc. Compositions and methods for production of immunoglobulins
PT2380592T (en) 2005-11-14 2018-06-06 Teva Pharmaceuticals Int Gmbh Antagonist antibody directed against calcitonin gene-related peptide
US8124103B2 (en) * 2006-02-13 2012-02-28 Fraunhofer Usa, Inc Influenza antigens, vaccine compositions, and related methods
WO2007095320A2 (en) * 2006-02-13 2007-08-23 Fraunhofer Usa, Inc. Hpv antigens, vaccine compositions, and related methods
WO2008048344A2 (en) * 2006-02-13 2008-04-24 Fraunhofer Usa, Inc. Bacillus anthracis antigens, vaccine compositions, and related methods
WO2007103290A2 (en) * 2006-03-03 2007-09-13 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Materials and methods directed to asparagine synthetase and asparaginase therapies
US8097425B2 (en) * 2006-03-10 2012-01-17 Tethys Bioscience, Inc. Multiplex protein fractionation
US20100278821A1 (en) * 2006-03-21 2010-11-04 The Regents Of The University Of California N-cadherin: target for cancer diagnosis and therapy
US8663635B2 (en) 2006-03-21 2014-03-04 The Regents Of The University Of California N-cadherin: target for cancer diagnosis and therapy
CA2646597A1 (en) * 2006-03-21 2007-09-27 The Regents Of The University Of California N-cadherin and ly6 e: targets for cancer diagnosis and therapy
EP2019684A4 (en) * 2006-04-20 2009-08-26 Jackson H M Found Military Med Methods and compositions based on shiga toxin type 1 protein
TWI429658B (en) 2006-06-07 2014-03-11 Bioalliance Cv Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
WO2008066691A2 (en) 2006-11-08 2008-06-05 Macrogenics West, Inc. Tes7 and antibodies that bind thereto
EP2144613B1 (en) 2006-12-29 2018-03-21 OstéoQC Inc. Methods of altering bone growth by administration of sost or wise antagonist or agonist
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US20100261640A1 (en) 2007-04-10 2010-10-14 Branco Luis M Soluble and membrane anchored forms of lassa virus subunit proteins
WO2008134643A2 (en) * 2007-04-28 2008-11-06 Fraunhofer Usa, Inc. Trypanosoma antigens, vaccine compositions, and related methods
CA2692933C (en) 2007-07-11 2016-10-18 Fraunhofer Usa, Inc. Yersinia pestis antigens, vaccine compositions, and related methods
EP2641618A3 (en) 2007-07-16 2013-10-23 Genentech, Inc. Humanized anti-CD79B antibodies and immunoconjugates and methods of use
SG183023A1 (en) 2007-07-16 2012-08-30 Genentech Inc Anti-cd79b antibodies and immunoconjugates and methods of use
JP2010534685A (en) * 2007-07-27 2010-11-11 ファセット バイオテック コーポレイション Pharmaceutical combination comprising a tyrosine kinase inhibitor and an antibody against integrin α5β1 (CD49E)
CN101918448A (en) 2007-12-18 2010-12-15 生物联合公司 Antibodies recognizing a carbohydrate containing epitope on CD-43 and CEA expressed on cancer cells and methods using same
IL295449A (en) 2008-01-31 2022-10-01 Genentech Inc Cycteine engineering anti-cd79b antibodies and antibody-drug conjugates
EP3045475B1 (en) 2008-04-02 2017-10-04 MacroGenics, Inc. Bcr-complex-specific antibodies and methods of using same
AU2009231570A1 (en) * 2008-04-04 2009-10-08 The Regents Of The University Of California Novel antibodies against cancer target block tumor growth, angiogenesis and metastasis
WO2009150623A1 (en) 2008-06-13 2009-12-17 Pfizer Inc Treatment of chronic prostatitis
TWI445716B (en) 2008-09-12 2014-07-21 Rinat Neuroscience Corp Pcsk9 antagonists
RU2518278C2 (en) 2008-09-19 2014-06-10 Пфайзер Инк. Stable liquid preparation of antibody
US8734803B2 (en) 2008-09-28 2014-05-27 Ibio Inc. Humanized neuraminidase antibody and methods of use thereof
US8703920B2 (en) * 2008-11-10 2014-04-22 The Regents Of The University Of California Fully human antibodies against N-cadherin
WO2010065544A2 (en) * 2008-12-01 2010-06-10 The Johns Hopkins University Diagnostic and treatment methods for cancer based on immune inhibitors
MX2011007866A (en) 2009-01-23 2011-10-06 T Of Military Medicine Inc The Henry M Jackson Foundation For The Advancemen Methods and compositions based on shiga toxin type 2 protein.
WO2010086828A2 (en) 2009-02-02 2010-08-05 Rinat Neuroscience Corporation Agonist anti-trkb monoclonal antibodies
CA2761310C (en) 2009-05-07 2017-02-28 Charles S. Craik Antibodies and methods of use thereof
WO2010146511A1 (en) 2009-06-17 2010-12-23 Pfizer Limited Treatment of overactive bladder
CA2765989C (en) 2009-06-18 2016-11-29 Pfizer Inc. Anti notch-1 antibodies
JP5883384B2 (en) 2009-08-13 2016-03-15 ザ ジョンズ ホプキンス ユニバーシティー How to regulate immune function
WO2011041391A1 (en) 2009-09-29 2011-04-07 Fraunhofer Usa, Inc. Influenza hemagglutinin antibodies, compositions, and related methods
US20110118178A1 (en) * 2009-11-13 2011-05-19 Sanofi-Aventis Deutschland Gmbh Method of treatment of diabetes type 2 comprising add-on therapy to insulin glargine and metformin
WO2011079283A1 (en) * 2009-12-23 2011-06-30 Bioalliance C.V. Anti-epcam antibodies that induce apoptosis of cancer cells and methods using same
AR080291A1 (en) 2010-02-24 2012-03-28 Rinat Neuroscience Corp ANTI-BODIES ANTAGONISTS ANTI RECEIVER OF IL-7 AND PROCEDURES
PE20130479A1 (en) 2010-03-04 2013-05-12 Macrogenics Inc ANTIBODIES REACTIVE WITH B7-H3, IMMULOGICALLY ACTIVE FRAGMENTS OF THE SAME AND USES OF THE SAME
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
KR20120138241A (en) 2010-03-11 2012-12-24 화이자 인코포레이티드 Antibodies with ph dependent antigen binding
RU2601297C2 (en) 2010-06-22 2016-10-27 Ридженерон Фармасьютикалз, Инк. Mice expressing an immunoglobulin hybrid light chain
US8747844B2 (en) 2010-07-30 2014-06-10 Saint Louis University Methods of treating pain
KR102434557B1 (en) * 2010-08-02 2022-08-23 리제너론 파아마슈티컬스, 인크. Mice that make binding proteins comprising vl domains
PL3333188T3 (en) 2010-08-19 2022-05-09 Zoetis Belgium S.A. Anti-ngf antibodies and their use
WO2012071216A2 (en) 2010-11-22 2012-05-31 Duke University Antibodies for tumor gangliosides
JP6016800B2 (en) 2010-12-15 2016-10-26 ワイス・エルエルシー Anti-notch1 antibody
SI2714733T1 (en) 2011-05-21 2019-06-28 Macrogenics, Inc. Cd3-binding molecules capable of binding to human and non-human cd3
JP5926374B2 (en) 2011-06-10 2016-05-25 メルサナ セラピューティクス,インコーポレイティド Protein-polymer-drug conjugate
MY170725A (en) 2011-09-09 2019-08-27 Univ Osaka Dengue-virus serotype neutralizing antibodies
BR112014011331A2 (en) 2011-11-11 2017-04-25 Rinat Neuroscience Corp trop-2 specific antibodies and their uses
BR112014015238B1 (en) 2011-12-20 2022-11-16 Regeneron Pharmaceuticals, Inc EX VIVO METHOD FOR PREPARING AN ANTIBODY THAT BINDS TO AN ANTIGEN OF INTEREST, INCLUDING IDENTIFYING NUCLEIC ACID SEQUENCES FROM MOUSE B LYMPHOCYTES GENETICALLY MODIFIED BY PLACING AN ADAM6 GENE
EP2794659A1 (en) 2011-12-22 2014-10-29 Rinat Neuroscience Corp. Human growth hormone receptor antagonist antibodies and methods of use thereof
WO2013093693A1 (en) 2011-12-22 2013-06-27 Rinat Neuroscience Corp. Staphylococcus aureus specific antibodies and uses thereof
US10114023B2 (en) 2012-04-18 2018-10-30 Massachusetts Institute Of Technology Method of enhancing the efficacy of anti-hepatocyte growth factor receptor breast cancer therapy by administering an inhibitor of menaINV
JP6629069B2 (en) 2012-06-06 2020-01-15 ゾエティス・エルエルシー Canine anti-NGF antibody and method thereof
AU2013286866B2 (en) 2012-07-02 2018-03-01 Commonwealth Scientific And Industrial Research Organization Paramyxovirus and methods of use
CA2889764C (en) 2012-11-01 2023-10-10 Martin Lipp An antibody that binds cd269 (bcma) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
BR112015010722A2 (en) 2012-11-09 2017-08-22 Pfizer SPECIFIC GROWTH FACTOR ANTIBODIES DERIVED FROM ISOFORM B PLATELETS AND COMPOSITIONS AND USES THEREOF
EP2916835A4 (en) 2012-11-12 2016-07-27 Redwood Bioscience Inc Compounds and methods for producing a conjugate
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
KR20150085064A (en) 2012-11-16 2015-07-22 더 리젠트스 오브 더 유니이버시티 오브 캘리포니아 Pictet-spengler ligation for protein chemical modification
US10226535B2 (en) 2012-12-10 2019-03-12 Mersana Therapeutics, Inc. Auristatin compounds and conjugates thereof
JP6334553B2 (en) 2012-12-10 2018-05-30 メルサナ セラピューティクス,インコーポレイティド Protein-polymer-drug conjugate
US9745558B2 (en) 2013-02-18 2017-08-29 Vegenics Pty Limited VEGFR-3 ligand binding molecules and uses thereof
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US9908938B2 (en) 2013-03-14 2018-03-06 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP3457138A3 (en) 2013-04-30 2019-06-19 Université de Montréal Novel biomarkers for acute myeloid leukemia
SG11201508264UA (en) 2013-05-07 2015-11-27 Rinat Neuroscience Corp Anti-glucagon receptor antibodies and methods of use thereof
JP6403668B2 (en) 2013-05-30 2018-10-10 国立大学法人千葉大学 Composition for treating inflammatory diseases comprising anti-myosin regulatory light chain polypeptide antibody
MX371455B (en) 2013-08-02 2020-01-28 Pfizer Anti-cxcr4 antibodies and antibody-drug conjugates.
WO2015050663A1 (en) 2013-10-01 2015-04-09 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of bim
ES2726850T3 (en) 2013-10-11 2019-10-09 Mersana Therapeutics Inc Protein-polymer-drug conjugates
CN105813655B (en) 2013-10-11 2022-03-15 阿萨纳生物科技有限责任公司 Protein-polymer-drug conjugates
KR102366076B1 (en) 2013-11-13 2022-02-21 화이자 인코포레이티드 Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
CA2927806C (en) 2013-11-27 2023-01-10 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
WO2015087187A1 (en) 2013-12-10 2015-06-18 Rinat Neuroscience Corp. Anti-sclerostin antibodies
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
ES2939760T3 (en) 2014-03-15 2023-04-26 Novartis Ag Cancer treatment using a chimeric receptor for antigens
US9896502B2 (en) 2014-03-21 2018-02-20 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
SG11201607015VA (en) 2014-03-21 2016-09-29 Regeneron Pharma V<sb>L</sb> ANTIGEN BINDING PROTEINS EXHIBITING DISTINCT BINDING CHARACTERISTICS
KR102601491B1 (en) 2014-03-21 2023-11-13 리제너론 파마슈티칼스 인코포레이티드 Non-human animals that make single domain binding proteins
RS61516B1 (en) 2014-04-30 2021-03-31 Pfizer Anti-ptk7 antibody-drug conjugates
WO2015179654A1 (en) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti b7-h1 antibodies
US10647768B2 (en) 2014-05-29 2020-05-12 Macrogenics, Inc. Multi-chain polypeptide-containing tri-specific binding molecules
EP3151830A4 (en) 2014-06-06 2018-02-07 Redwood Bioscience, Inc. Anti-her2 antibody-maytansine conjugates and methods of use thereof
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
JP2017528433A (en) 2014-07-21 2017-09-28 ノバルティス アーゲー Low immunoenhancing dose of mTOR inhibitor and CAR combination
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
WO2016034968A1 (en) 2014-09-02 2016-03-10 Pfizer Inc. Therapeutic antibody
ES2796903T3 (en) 2014-09-23 2020-11-30 Hoffmann La Roche Procedure for the use of anti-CD79b immunoconjugates
UY36316A (en) 2014-09-26 2016-04-29 Macrogenics Inc MONOVALENT BIESPECFIC DIACUMS THAT ARE ABLE TO JOIN CD19 AND CD3, AND USES OF THE SAME
SG11201702544WA (en) 2014-09-29 2017-04-27 Univ Duke Bispecific molecules comprising an hiv-1 envelope targeting arm
CN107073113A (en) 2014-10-18 2017-08-18 辉瑞大药厂 Anti- IL 7R antibody compositions
CA2959821A1 (en) 2014-10-24 2016-04-28 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for inducing phagocytosis of mhc class i positive cells and countering anti-cd47/sirpa resistance
WO2016081455A1 (en) 2014-11-17 2016-05-26 Pelican Therapeutics, Inc. Human tnfrsf25 antibody
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
US20170360929A1 (en) 2014-12-23 2017-12-21 Pfizer Inc. Stable aqueous antibody formulation for anti tnf alpha antibodies
KR20170105622A (en) 2015-01-26 2017-09-19 마크로제닉스, 인크. A multivalent molecule comprising a DR5-binding domain
RU2699544C2 (en) 2015-01-28 2019-09-06 Пфайзер Инк. Stable aqueous anti-vascular endothelial growth factor (vegf) antibody composition
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
CN107438622A (en) 2015-03-19 2017-12-05 瑞泽恩制药公司 Non-human animal of the selection with reference to the light chain variable district of antigen
BR112017019785B1 (en) 2015-04-13 2022-11-16 Pfizer Inc BISPECIFIC ANTIBODY, ITS USE AND PHARMACEUTICAL COMPOSITION
US20180298068A1 (en) 2015-04-23 2018-10-18 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
TW201709929A (en) 2015-06-12 2017-03-16 宏觀基因股份有限公司 Combination therapy for the treatment of cancer
WO2017015334A1 (en) 2015-07-21 2017-01-26 Saint Louis University Compositions and methods for diagnosing and treating endometriosis-related infertility
WO2017015619A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
LT3328419T (en) 2015-07-30 2021-11-10 Macrogenics, Inc. Pd-1-binding molecules and methods of use thereof
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
MX2018004177A (en) 2015-10-08 2018-09-11 Macrogenics Inc Combination therapy for the treatment of cancer.
US10138298B2 (en) 2015-10-23 2018-11-27 The Regents Of The University Of California Anti-IL-2 antibodies and compositions and uses thereof
WO2017075045A2 (en) 2015-10-30 2017-05-04 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
WO2017079419A1 (en) 2015-11-05 2017-05-11 The Regents Of The University Of California Cells labelled with lipid conjugates and methods of use thereof
JP7118887B2 (en) 2015-11-23 2022-08-16 ノバルティス アーゲー Optimized lentiviral transfer vectors and their uses
EP3389714A4 (en) 2015-12-14 2019-11-13 MacroGenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
RU2018127657A (en) 2015-12-30 2020-01-31 Новартис Аг TYPES OF THERAPY BASED ON IMMUNO EFFECTIVE CELLS WITH IMPROVED EFFICIENCY
CN108713026B (en) 2016-01-08 2023-01-06 美国全心医药生技股份有限公司 Tetravalent anti-PSGL-1 antibodies and uses thereof
TW201936640A (en) 2016-01-21 2019-09-16 美商輝瑞股份有限公司 Antibodies specific for epidermal growth factor receptor variant III and their uses
TW201730212A (en) 2016-02-17 2017-09-01 宏觀基因股份有限公司 ROR1-binding molecules, and methods of use thereof
US10443054B2 (en) 2016-03-06 2019-10-15 Massachusetts Institute Of Technology Methods for identifying and treating invasive/metastatic breast cancers
EP3432924A1 (en) 2016-03-23 2019-01-30 Novartis AG Cell secreted minibodies and uses thereof
IL262396B2 (en) 2016-04-15 2023-09-01 Macrogenics Inc Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
JP2019527537A (en) 2016-06-07 2019-10-03 マックス−デルブリュック−セントルム フュール モレキュラー メディツィン イン デア ヘルムホルツ−ゲマインシャフト Chimeric antigen receptor and CAR-T cell binding to BCMA
US10005843B2 (en) 2016-06-09 2018-06-26 Pelican Therapeutics, Inc. Anti-TNFRSF25 antibodies
CA3040343A1 (en) 2016-10-19 2018-04-26 California Institute For Biomedical Research Chimeric antigen receptor effector cell switches with humanized targeting moieties and/or optimized chimeric antigen receptor interacting domains and uses thereof
WO2018106712A1 (en) 2016-12-05 2018-06-14 The Administrators Of The Tulane Educational Fund Arenavirus monoclonal antibodies and uses
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
PL3558391T3 (en) 2016-12-23 2022-05-16 Immunogen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
EP3558368A4 (en) 2016-12-23 2020-12-30 MacroGenics, Inc. Adam9-binding molecules, and methods of use thereof
ES2912408T3 (en) 2017-01-26 2022-05-25 Novartis Ag CD28 compositions and methods for therapy with chimeric receptors for antigens
WO2018139608A1 (en) 2017-01-27 2018-08-02 株式会社A-Clip研究所 Preventive and/or therapeutic agent for infectious diseases or inflammatory diseases
US20190375815A1 (en) 2017-01-31 2019-12-12 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
CN110325209A (en) 2017-02-24 2019-10-11 宏观基因有限公司 CD137 and the bi-specific binding molecule of tumour antigen and application thereof can be combined
EP3589647A1 (en) 2017-02-28 2020-01-08 Novartis AG Shp inhibitor compositions and uses for chimeric antigen receptor therapy
EP3589657A1 (en) 2017-03-03 2020-01-08 Rinat Neuroscience Corp. Anti-gitr antibodies and methods of use thereof
CA3059542A1 (en) 2017-04-12 2018-10-18 Pfizer Inc. Antibodies having conditional affinity and methods of use thereof
MX2019014465A (en) 2017-06-02 2020-01-23 Pfizer Antibodies specific for flt3 and their uses.
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
JP7395465B2 (en) 2017-08-23 2023-12-11 マックス-デルブリュック-セントルム フュール モレキュラー メディツィン イン デア ヘルムホルツ-ゲマインシャフト Chimeric antigen receptor and CAR-T cells that bind to CXCR5
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
RU2020116579A (en) 2017-10-25 2021-11-25 Новартис Аг METHODS FOR OBTAINING CELLS EXPRESSING A CHIMERIC ANTIGENIC RECEPTOR
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
SG11202003044SA (en) * 2017-12-05 2020-04-29 Regeneron Pharma Non-human animals having an engineered immunoglobulin lambda light chain and uses thereof
RU2020122822A (en) 2017-12-12 2022-01-13 Макродженикс, Инк. BISPECIFIC CD16 BINDING MOLECULES AND THEIR USE IN THE TREATMENT OF DISEASES
JP2021511811A (en) 2018-02-01 2021-05-13 ファイザー・インク CD70-specific antibodies and their use
WO2019152742A1 (en) 2018-02-01 2019-08-08 Pfizer Inc. Chimeric antigen receptors targeting cd70
JP7337079B2 (en) 2018-02-15 2023-09-01 マクロジェニクス,インコーポレーテッド Mutant CD3 binding domains and their use in combination therapy for the treatment of disease
WO2019166946A1 (en) 2018-02-28 2019-09-06 Pfizer Inc. Il-15 variants and uses thereof
EP3765499A1 (en) 2018-03-12 2021-01-20 Zoetis Services LLC Anti-ngf antibodies and methods thereof
WO2019201995A1 (en) 2018-04-20 2019-10-24 Medizinische Hochschule Hannover Chimeric antigen receptor and car-t cells that bind a herpes virus antigen
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
EP3788369A1 (en) 2018-05-01 2021-03-10 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
TWI816396B (en) 2018-05-23 2023-09-21 美商輝瑞大藥廠 Antibodies specific for gucy2c and uses thereof
MX2020012539A (en) 2018-05-23 2021-02-16 Pfizer Antibodies specific for cd3 and uses thereof.
EP3801769A1 (en) 2018-05-25 2021-04-14 Novartis AG Combination therapy with chimeric antigen receptor (car) therapies
EP3802825A1 (en) 2018-06-08 2021-04-14 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
WO2019234680A1 (en) 2018-06-08 2019-12-12 Pfizer Inc. Methods of treating iron metabolic disease with a neutralizing antibody binding erhythroferrone
EP3814378A1 (en) 2018-06-26 2021-05-05 ImmunoGen, Inc. Immunoconjugates targeting adam9 and methods of use thereof
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
CN110760002A (en) * 2018-07-25 2020-02-07 南京金斯瑞生物科技有限公司 Humanized anti-human CTLA4 monoclonal antibody and preparation method and application thereof
EP3892639A4 (en) 2018-12-07 2022-08-24 Jiangsu Hengrui Medicine Co., Ltd. Cd3 antibody and pharmaceutical use thereof
BR112021011874A2 (en) 2018-12-20 2021-09-08 Novartis Ag DOSAGE SCHEME AND PHARMACEUTICAL COMBINATION INCLUDING DERIVATIVES OF 3-(1-OXOISOINDOLIN-2-YL)PIPERIDINE-2,6-DIONE
KR20210129672A (en) 2019-02-15 2021-10-28 노파르티스 아게 Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
MX2021009763A (en) 2019-02-15 2021-09-08 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof.
KR20210141948A (en) 2019-03-19 2021-11-23 앨버트 아인슈타인 컬리지 오브 메디신 Monoclonal Antibodies for Prevention and Treatment of Herpes Simplex Virus Infection
US20220154191A1 (en) 2019-03-25 2022-05-19 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Enhancement of cytolytic t-cell activity by inhibiting ebag9
CA3165399A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021181233A2 (en) 2020-03-09 2021-09-16 Pfizer Inc. Fusion proteins and uses thereof
WO2021207348A1 (en) 2020-04-07 2021-10-14 Albert Einstein College Of Medicine Method of treating and preventing ocular disease with hsv-2 delta gd
IL298473A (en) 2020-06-11 2023-01-01 Novartis Ag Zbtb32 inhibitors and uses thereof
BR112022026202A2 (en) 2020-06-23 2023-01-17 Novartis Ag DOSAGE REGIMEN COMPRISING 3-(1-OXOISOINDOLIN-2-IL)PIPERIDINE-2,6-DIONE DERIVATIVES
EP4175650A1 (en) 2020-07-06 2023-05-10 Kiromic BioPharma, Inc. Mesothelin isoform binding molecules and chimeric pd1 receptor molecules, cells containing the same and uses thereof
BR112023000650A2 (en) 2020-07-14 2023-01-31 Pfizer RECOMBINANT VACCINIA VIRUS
CA3189590A1 (en) 2020-07-17 2022-01-20 Pfizer Inc. Therapeutic antibodies and their uses
CN116134027A (en) 2020-08-03 2023-05-16 诺华股份有限公司 Heteroaryl-substituted 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione derivatives and uses thereof
US20240009310A1 (en) 2020-08-24 2024-01-11 Charité - Universitätsmedizin Berlin A CHIMERIC ANTIGEN RECEPTOR CONSTRUCT ENCODING A CHECKPOINT INHIBITORY MOLECULE AND AN IMMUNE STIMULATORY CYTOKINE AND CAR-EXPRESSING CELLS RECOGNIZING CD44v6
US20230321242A1 (en) 2020-08-24 2023-10-12 Charité - Universitätsmedizin Berlin Chimeric antigen receptor (car)-expressing cells recognizing cea
CA3202233A1 (en) 2020-11-18 2022-05-27 Kiromic BioPharma, Inc. Gamma-delta t cell manufacturing processes and chimeric pd1 receptor molecules
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS60258128A (en) * 1984-06-06 1985-12-20 Kyowa Hakko Kogyo Co Ltd Rat monoclonal antibody
JPS6147500A (en) * 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) * 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) * 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
GB8607679D0 (en) * 1986-03-27 1986-04-30 Winter G P Recombinant dna product
NO873164L (en) * 1986-07-30 1988-02-01 Teijin Ltd MUSEUM-HUMAN CHEMICAL ANTIBODIES.
IL84285A (en) * 1986-10-27 1993-03-15 Int Genetic Engineering Chimeric antibody with specificity to human tumor antigen
CA1341374C (en) * 1986-11-13 2002-07-09 Alan N. Houghton Compositions and method for treatment of cancer using monoclonal antibody against gd3 ganglioside together with il-2
US4849509A (en) * 1987-02-20 1989-07-18 The Wistar Institute Monoclonal antibodies against melanoma-associated antigens and hybrid cell lines producing these antibodies
FI884924A (en) * 1987-10-28 1989-04-29 Oncogen HUMANIMMUGLOBULIN SOM PRODUCERATS MED HYBRID-DNA-TEKNIK.
JP2706777B2 (en) * 1988-02-19 1998-01-28 メクト株式会社 Monoclonal antibody recognizing non-natural GD
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
ES2098304T3 (en) * 1990-11-30 1997-05-01 Kyowa Hakko Kogyo Kk MONOCLONAL ANTIBODIES AGAINST THE GLYCOLIPIDIC CARBON HYDRATE CHAINS.
AU669124B2 (en) 1991-09-18 1996-05-30 Kyowa Hakko Kirin Co., Ltd. Process for producing humanized chimera antibody

Also Published As

Publication number Publication date
AU691116B2 (en) 1998-05-07
DK0533199T3 (en) 2003-08-25
US6437098B1 (en) 2002-08-20
EP1013761A3 (en) 2004-11-03
AU669124B2 (en) 1996-05-30
ES2195995T3 (en) 2003-12-16
US20030095964A1 (en) 2003-05-22
US5866692A (en) 1999-02-02
EP0533199B1 (en) 2003-05-02
EP1013761A2 (en) 2000-06-28
US20020026036A1 (en) 2002-02-28
EP0533199A2 (en) 1993-03-24
EP0533199A3 (en) 1993-10-06
ATE239078T1 (en) 2003-05-15
US5807548A (en) 1998-09-15
JP3440104B2 (en) 2003-08-25
EP1013761B1 (en) 2007-08-08
US20030166876A1 (en) 2003-09-04
AU2458192A (en) 1993-03-25
US7045129B2 (en) 2006-05-16
DE69233027T2 (en) 2004-03-11
PT533199E (en) 2003-09-30
US20060058512A1 (en) 2006-03-16
US20070073043A1 (en) 2007-03-29
CA2078539A1 (en) 1993-03-19
DE69233706D1 (en) 2007-09-20
DE69233027D1 (en) 2003-06-05
US6495666B2 (en) 2002-12-17
US5750078A (en) 1998-05-12
ATE369424T1 (en) 2007-08-15
AU6069496A (en) 1996-10-24
JPH05304989A (en) 1993-11-19
US20060057139A1 (en) 2006-03-16
US6965024B2 (en) 2005-11-15
DE69233706T2 (en) 2008-05-21

Similar Documents

Publication Publication Date Title
CA2078539C (en) Process for producing humanized chimera antibody
EP0598998B1 (en) Humanized antibodies reacting with the ganglioside GM2
US5354847A (en) Chimeric antibody with specificity to human tumor antigen
US6042828A (en) Humanized antibodies to ganglioside GM2
CA1336826C (en) Chimeric antibody with specificity to human b cell surface antigen
US6893625B1 (en) Chimeric antibody with specificity to human B cell surface antigen
CA2280875C (en) Remedies for lymphocytic tumors
US5939532A (en) Humanized antibodies to ganglioside GM2
JP3565572B2 (en) Humanized antibodies
Nakamura et al. Humanized antibodies to ganglioside GM 2

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry